Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Med Chem ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009041

ABSTRACT

Identifying promising chemical starting points for small molecule inhibitors of active, GTP-loaded KRAS "on" remains of great importance to clinical oncology and represents a significant challenge in medicinal chemistry. Here, we describe broadly applicable learnings from a KRAS hit finding campaign: While we initially identified KRAS inhibitors in a biochemical high-throughput screen, we later discovered that compound potencies were all but assay artifacts linked to metal salts interfering with KRAS AlphaScreen assay technology. The source of the apparent biochemical KRAS inhibition was ultimately traced to unavoidable palladium impurities from chemical synthesis. This discovery led to the development of a Metal Ion Interference Set (MIIS) for up-front assay development and testing. Profiling of the MIIS across 74 assays revealed a reduced interference liability of label-free biophysical assays and, as a result, provided general estimates for luminescence- and fluorescence-based assay susceptibility to metal salt interference.

2.
ACS Chem Biol ; 19(7): 1484-1494, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38958654

ABSTRACT

Targeted protein degradation has recently emerged as a novel option in drug discovery. Natural protein half-life is expected to affect the efficacy of degrading agents, but to what extent it influences target protein degradation has not been systematically explored. Using simple mathematical modeling of protein degradation, we find that the natural half-life of a target protein has a dramatic effect on the level of protein degradation induced by a degrader agent which can pose significant hurdles to screening efforts. Moreover, we show that upon screening for degraders of short-lived proteins, agents that stall protein synthesis, such as GSPT1 degraders and generally cytotoxic compounds, deceptively appear as protein-degrading agents. This is exemplified by the disappearance of short-lived proteins such as MCL1 and MDM2 upon GSPT1 degradation and upon treatment with cytotoxic agents such as doxorubicin. These findings have implications for target selection as well as for the type of control experiments required to conclude that a novel agent works as a bona fide targeted protein degrader.


Subject(s)
Proteolysis , Humans , Proto-Oncogene Proteins c-mdm2/metabolism , Half-Life , Doxorubicin/pharmacology , Doxorubicin/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proteins/metabolism , Proteins/chemistry
3.
ChemMedChem ; 18(6): e202200686, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36649575

ABSTRACT

The bromodomain and PHD-finger containing transcription factor (BPTF) is part of the nucleosome remodeling factor (NURF) complex and has been implicated in multiple cancer types. Here, we report the discovery of a potent and selective chemical probe targeting the bromodomain of BPTF with an attractive pharmacokinetic profile enabling cellular and in vivo experiments in mice. Microarray-based transcriptomics in presence of the probe in two lung cancer cell lines revealed only minor effects on the transcriptome. Profiling against a panel of cancer cell lines revealed that the antiproliferative effect does not correlate with BPTF dependency score in depletion screens. Both observations and the multi-domain architecture of BPTF suggest that depleting the protein by proteolysis targeting chimeras (PROTACs) could be a promising strategy to target cancer cell proliferation. We envision that the presented chemical probe and the related negative control will enable the research community to further explore scientific hypotheses with respect to BPTF bromodomain inhibition.


Subject(s)
Lung Neoplasms , Transcription Factors , Animals , Mice , Cell Proliferation , Gene Expression Regulation , Nuclear Proteins/metabolism , Transcription Factors/metabolism
4.
Nat Commun ; 13(1): 5969, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36216795

ABSTRACT

Targeted protein degradation offers an alternative modality to classical inhibition and holds the promise of addressing previously undruggable targets to provide novel therapeutic options for patients. Heterobifunctional molecules co-recruit a target protein and an E3 ligase, resulting in ubiquitylation and proteosome-dependent degradation of the target. In the clinic, the oral route of administration is the option of choice but has only been achieved so far by CRBN- recruiting bifunctional degrader molecules. We aimed to achieve orally bioavailable molecules that selectively degrade the BAF Chromatin Remodelling complex ATPase SMARCA2 over its closely related paralogue SMARCA4, to allow in vivo evaluation of the synthetic lethality concept of SMARCA2 dependency in SMARCA4-deficient cancers. Here we outline structure- and property-guided approaches that led to orally bioavailable VHL-recruiting degraders. Our tool compound, ACBI2, shows selective degradation of SMARCA2 over SMARCA4 in ex vivo human whole blood assays and in vivo efficacy in SMARCA4-deficient cancer models. This study demonstrates the feasibility for broadening the E3 ligase and physicochemical space that can be utilised for achieving oral efficacy with bifunctional molecules.


Subject(s)
Adenosine Triphosphatases , Transcription Factors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteolysis , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
J Med Chem ; 65(21): 14614-14629, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36300829

ABSTRACT

Activating mutations in KRAS are the most frequent oncogenic alterations in cancer. The oncogenic hotspot position 12, located at the lip of the switch II pocket, offers a covalent attachment point for KRASG12C inhibitors. To date, KRASG12C inhibitors have been discovered by first covalently binding to the cysteine at position 12 and then optimizing pocket binding. We report on the discovery of the in vivo active KRASG12C inhibitor BI-0474 using a different approach, in which small molecules that bind reversibly to the switch II pocket were identified and then optimized for non-covalent binding using structure-based design. Finally, the Michael acceptor containing warhead was attached. Our approach offers not only an alternative approach to discovering KRASG12C inhibitors but also provides a starting point for the discovery of inhibitors against other oncogenic KRAS mutants.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Genes, ras , Mutation , Neoplasms/genetics , Cysteine
6.
Nat Cancer ; 3(7): 821-836, 2022 07.
Article in English | MEDLINE | ID: mdl-35883003

ABSTRACT

Oncogenic alterations in human epidermal growth factor receptor 2 (HER2) occur in approximately 2% of patients with non-small cell lung cancer and predominantly affect the tyrosine kinase domain and cluster in exon 20 of the ERBB2 gene. Most clinical-grade tyrosine kinase inhibitors are limited by either insufficient selectivity against wild-type (WT) epidermal growth factor receptor (EGFR), which is a major cause of dose-limiting toxicity or by potency against HER2 exon 20 mutant variants. Here we report the discovery of covalent tyrosine kinase inhibitors that potently inhibit HER2 exon 20 mutants while sparing WT EGFR, which reduce tumor cell survival and proliferation in vitro and result in regressions in preclinical xenograft models of HER2 exon 20 mutant non-small cell lung cancer, concomitant with inhibition of downstream HER2 signaling. Our results suggest that HER2 exon 20 insertion-driven tumors can be effectively treated by a potent and highly selective HER2 inhibitor while sparing WT EGFR, paving the way for clinical translation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Exons/genetics , Genes, erbB-2 , Humans , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/genetics
7.
J Med Chem ; 64(10): 6569-6580, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33719426

ABSTRACT

KRAS, the most common oncogenic driver in human cancers, is controlled and signals primarily through protein-protein interactions (PPIs). The interaction between KRAS and SOS1, crucial for the activation of KRAS, is a typical, challenging PPI with a large contact surface area and high affinity. Here, we report that the addition of only one atom placed between Y884SOS1 and A73KRAS is sufficient to convert SOS1 activators into SOS1 inhibitors. We also disclose the discovery of BI-3406. Combination with the upstream EGFR inhibitor afatinib shows in vivo efficacy against KRASG13D mutant colorectal tumor cells, demonstrating the utility of BI-3406 to probe SOS1 biology. These findings challenge the dogma that large molecules are required to disrupt challenging PPIs. Instead, a "foot in the door" approach, whereby single atoms or small functional groups placed between key PPI interactions, can lead to potent inhibitors even for challenging PPIs such as SOS1-KRAS.


Subject(s)
Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/metabolism , Afatinib/chemistry , Afatinib/metabolism , Afatinib/therapeutic use , Allosteric Regulation/drug effects , Binding Sites , Catalytic Domain , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Interaction Maps/drug effects , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Quinazolines/chemistry , Quinazolines/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , SOS1 Protein/agonists , SOS1 Protein/antagonists & inhibitors , SOS1 Protein/genetics
8.
Cancer Discov ; 11(1): 142-157, 2021 01.
Article in English | MEDLINE | ID: mdl-32816843

ABSTRACT

KRAS is the most frequently mutated driver of pancreatic, colorectal, and non-small cell lung cancers. Direct KRAS blockade has proved challenging, and inhibition of a key downstream effector pathway, the RAF-MEK-ERK cascade, has shown limited success because of activation of feedback networks that keep the pathway in check. We hypothesized that inhibiting SOS1, a KRAS activator and important feedback node, represents an effective approach to treat KRAS-driven cancers. We report the discovery of a highly potent, selective, and orally bioavailable small-molecule SOS1 inhibitor, BI-3406, that binds to the catalytic domain of SOS1, thereby preventing the interaction with KRAS. BI-3406 reduces formation of GTP-loaded RAS and limits cellular proliferation of a broad range of KRAS-driven cancers. Importantly, BI-3406 attenuates feedback reactivation induced by MEK inhibitors and thereby enhances sensitivity of KRAS-dependent cancers to MEK inhibition. Combined SOS1 and MEK inhibition represents a novel and effective therapeutic concept to address KRAS-driven tumors. SIGNIFICANCE: To date, there are no effective targeted pan-KRAS therapies. In-depth characterization of BI-3406 activity and identification of MEK inhibitors as effective combination partners provide an attractive therapeutic concept for the majority of KRAS-mutant cancers, including those fueled by the most prevalent mutant KRAS oncoproteins, G12D, G12V, G12C, and G13D.See related commentary by Zhao et al., p. 17.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins p21(ras) , Cell Line, Tumor , Humans , Mitogen-Activated Protein Kinase Kinases , Mutation , Nucleotides , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics
10.
Mol Cancer Ther ; 19(4): 1018-1030, 2020 04.
Article in English | MEDLINE | ID: mdl-32024684

ABSTRACT

Natural killer (NK) cells play a pivotal role in controlling cancer. Multiple extracellular receptors and internal signaling nodes tightly regulate NK activation. Cyclin-dependent kinases of the mediator complex (CDK8 and CDK19) were described as a signaling intermediates in NK cells. Here, we report for the first time the development and use of CDK8/19 inhibitors to suppress phosphorylation of STAT1S727 in NK cells and to augment the production of the cytolytic molecules perforin and granzyme B (GZMB). Functionally, this resulted in enhanced NK-cell-mediated lysis of primary leukemia cells. Treatment with the CDK8/19 inhibitor BI-1347 increased the response rate and survival of mice bearing melanoma and breast cancer xenografts. In addition, CDK8/19 inhibition augmented the antitumoral activity of anti-PD-1 antibody and SMAC mimetic therapy, both agents that promote T-cell-mediated antitumor immunity. Treatment with the SMAC mimetic compound BI-8382 resulted in an increased number of NK cells infiltrating EMT6 tumors. Combination of the CDK8/19 inhibitor BI-1347, which augments the amount of degranulation enzymes, with the SMAC mimetic BI-8382 resulted in increased survival of mice carrying the EMT6 breast cancer model. The observed survival benefit was dependent on an intermittent treatment schedule of BI-1347, suggesting the importance of circumventing a hyporesponsive state of NK cells. These results suggest that CDK8/19 inhibitors can be combined with modulators of the adaptive immune system to inhibit the growth of solid tumors, independent of their activity on cancer cells, but rather through promoting NK-cell function.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/drug therapy , Melanoma, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Proliferation , Cytotoxicity, Immunologic/immunology , Female , Humans , Killer Cells, Natural/drug effects , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Melanoma, Experimental/enzymology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Phosphorylation , STAT1 Transcription Factor/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
J Med Chem ; 62(22): 10272-10293, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31689114

ABSTRACT

The epidermal growth factor receptor (EGFR), when carrying an activating mutation like del19 or L858R, acts as an oncogenic driver in a subset of lung tumors. While tumor responses to tyrosine kinase inhibitors (TKIs) are accompanied by marked tumor shrinkage, the response is usually not durable. Most patients relapse within two years of therapy often due to acquisition of an additional mutation in EGFR kinase domain that confers resistance to TKIs. Crucially, oncogenic EGFR harboring both resistance mutations, T790M and C797S, can no longer be inhibited by currently approved EGFR TKIs. Here, we describe the discovery of BI-4020, which is a noncovalent, wild-type EGFR sparing, macrocyclic TKI. BI-4020 potently inhibits the above-described EGFR variants and induces tumor regressions in a cross-resistant EGFRdel19 T790M C797S xenograft model. Key was the identification of a highly selective but moderately potent benzimidazole followed by complete rigidification of the molecule through macrocyclization.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Crystallography, X-Ray , Cyclization , Entropy , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , Female , Hepatocytes , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Mutation , Protein Conformation , Protein Kinase Inhibitors/pharmacokinetics , Structure-Activity Relationship , Xenograft Model Antitumor Assays
12.
J Med Chem ; 62(17): 7976-7997, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31365252

ABSTRACT

Phosphoglycerate dehydrogenase (PHGDH) is known to be the rate-limiting enzyme in the serine synthesis pathway in humans. It converts glycolysis-derived 3-phosphoglycerate to 3-phosphopyruvate in a co-factor-dependent oxidation reaction. Herein, we report the discovery of BI-4916, a prodrug of the co-factor nicotinamide adenine dinucleotide (NADH/NAD+)-competitive PHGDH inhibitor BI-4924, which has shown high selectivity against the majority of other dehydrogenase targets. Starting with a fragment-based screening, a subsequent hit optimization using structure-based drug design was conducted to deliver a single-digit nanomolar lead series and to improve potency by 6 orders of magnitude. To this end, an intracellular ester cleavage mechanism of the ester prodrug was utilized to achieve intracellular enrichment of the actual carboxylic acid based drug and thus overcome high cytosolic levels of the competitive cofactors NADH/NAD+.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Serine/antagonists & inhibitors , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indoles/chemical synthesis , Indoles/chemistry , Models, Molecular , Molecular Structure , Phosphoglycerate Dehydrogenase/metabolism , Serine/biosynthesis , Structure-Activity Relationship
13.
Nat Chem Biol ; 15(8): 846, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31267096

ABSTRACT

In the version of this article originally published, several lines of text in the last paragraph of the right column on page 1 of the PDF were transposed into the bottom paragraph of the left column. The affected text of the left column should read "The ATP-dependent activities of the BAF (SWI/SNF) chromatin remodeling complexes affect the positioning of nucleosomes on DNA and thereby many cellular processes related to chromatin structure, including transcription, DNA repair and decatenation of chromosomes during mitosis12,13." The affected text of the right column should read "SMARCA2/4BD inhibitors are thus precluded from use for the treatment of SMARCA4 mutant cancers but could provide attractive ligands for PROTAC conjugation. Small molecules binding to other bromodomains have been successfully converted into PROTACs by conjugating them with structures capable of binding to the E3 ligases von Hippel-Lindau (VHL) or cereblon5,6,10,11,25,26,27." The errors have been corrected in the PDF version of the paper.

14.
Nat Chem Biol ; 15(8): 822-829, 2019 08.
Article in English | MEDLINE | ID: mdl-31285596

ABSTRACT

Here, we report the fragment-based discovery of BI-9321, a potent, selective and cellular active antagonist of the NSD3-PWWP1 domain. The human NSD3 protein is encoded by the WHSC1L1 gene located in the 8p11-p12 amplicon, frequently amplified in breast and squamous lung cancer. Recently, it was demonstrated that the PWWP1 domain of NSD3 is required for the viability of acute myeloid leukemia cells. To further elucidate the relevance of NSD3 in cancer biology, we developed a chemical probe, BI-9321, targeting the methyl-lysine binding site of the PWWP1 domain with sub-micromolar in vitro activity and cellular target engagement at 1 µM. As a single agent, BI-9321 downregulates Myc messenger RNA expression and reduces proliferation in MOLM-13 cells. This first-in-class chemical probe BI-9321, together with the negative control BI-9466, will greatly facilitate the elucidation of the underexplored biological function of PWWP domains.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , CRISPR-Cas Systems , Cell Line , Cell Proliferation/drug effects , Cell Survival , Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
15.
Proc Natl Acad Sci U S A ; 116(32): 15823-15829, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31332011

ABSTRACT

The 3 human RAS genes, KRAS, NRAS, and HRAS, encode 4 different RAS proteins which belong to the protein family of small GTPases that function as binary molecular switches involved in cell signaling. Activating mutations in RAS are among the most common oncogenic drivers in human cancers, with KRAS being the most frequently mutated oncogene. Although KRAS is an excellent drug discovery target for many cancers, and despite decades of research, no therapeutic agent directly targeting RAS has been clinically approved. Using structure-based drug design, we have discovered BI-2852 (1), a KRAS inhibitor that binds with nanomolar affinity to a pocket, thus far perceived to be "undruggable," between switch I and II on RAS; 1 is mechanistically distinct from covalent KRASG12C inhibitors because it binds to a different pocket present in both the active and inactive forms of KRAS. In doing so, it blocks all GEF, GAP, and effector interactions with KRAS, leading to inhibition of downstream signaling and an antiproliferative effect in the low micromolar range in KRAS mutant cells. These findings clearly demonstrate that this so-called switch I/II pocket is indeed druggable and provide the scientific community with a chemical probe that simultaneously targets the active and inactive forms of KRAS.


Subject(s)
Drug Discovery , Pharmaceutical Preparations/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Guanosine Triphosphate/metabolism , Humans , Models, Molecular , Nanoparticles/chemistry
16.
Nat Chem Biol ; 15(7): 672-680, 2019 07.
Article in English | MEDLINE | ID: mdl-31178587

ABSTRACT

Targeting subunits of BAF/PBAF chromatin remodeling complexes has been proposed as an approach to exploit cancer vulnerabilities. Here, we develop proteolysis targeting chimera (PROTAC) degraders of the BAF ATPase subunits SMARCA2 and SMARCA4 using a bromodomain ligand and recruitment of the E3 ubiquitin ligase VHL. High-resolution ternary complex crystal structures and biophysical investigation guided rational and efficient optimization toward ACBI1, a potent and cooperative degrader of SMARCA2, SMARCA4 and PBRM1. ACBI1 induced anti-proliferative effects and cell death caused by SMARCA2 depletion in SMARCA4 mutant cancer cells, and in acute myeloid leukemia cells dependent on SMARCA4 ATPase activity. These findings exemplify a successful biophysics- and structure-based PROTAC design approach to degrade high profile drug targets, and pave the way toward new therapeutics for the treatment of tumors sensitive to the loss of BAF complex ATPases.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Molecular Structure , Nuclear Proteins/metabolism
17.
Oncogene ; 37(20): 2687-2701, 2018 05.
Article in English | MEDLINE | ID: mdl-29491412

ABSTRACT

Bromodomain and extra-terminal (BET) protein inhibitors have been reported as treatment options for acute myeloid leukemia (AML) in preclinical models and are currently being evaluated in clinical trials. This work presents a novel potent and selective BET inhibitor (BI 894999), which has recently entered clinical trials (NCT02516553). In preclinical studies, this compound is highly active in AML cell lines, primary patient samples, and xenografts. HEXIM1 is described as an excellent pharmacodynamic biomarker for target engagement in tumors as well as in blood. Mechanistic studies show that BI 894999 targets super-enhancer-regulated oncogenes and other lineage-specific factors, which are involved in the maintenance of the disease state. BI 894999 is active as monotherapy in AML xenografts, and in addition leads to strongly enhanced antitumor effects in combination with CDK9 inhibitors. This treatment combination results in a marked decrease of global p-Ser2 RNA polymerase II levels and leads to rapid induction of apoptosis in vitro and in vivo. Together, these data provide a strong rationale for the clinical evaluation of BI 894999 in AML.


Subject(s)
Antineoplastic Agents/administration & dosage , Enhancer Elements, Genetic/drug effects , Flavonoids/administration & dosage , Gene Expression Profiling/methods , Leukemia, Myeloid, Acute/drug therapy , Piperidines/administration & dosage , Proteins/antagonists & inhibitors , Pyrazines/administration & dosage , Triazoles/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Down-Regulation , Drug Synergism , Drug Therapy, Combination , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Piperidines/pharmacology , Pyrazines/pharmacology , RNA Polymerase II/metabolism , RNA-Binding Proteins/genetics , Transcription Factors , Triazoles/pharmacology , Xenograft Model Antitumor Assays
18.
Cell Rep ; 20(12): 2860-2875, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930682

ABSTRACT

The transcription factor BCL6 is a known driver of oncogenesis in lymphoid malignancies, including diffuse large B cell lymphoma (DLBCL). Disruption of its interaction with transcriptional repressors interferes with the oncogenic effects of BCL6. We used a structure-based drug design to develop highly potent compounds that block this interaction. A subset of these inhibitors also causes rapid ubiquitylation and degradation of BCL6 in cells. These compounds display significantly stronger induction of expression of BCL6-repressed genes and anti-proliferative effects than compounds that merely inhibit co-repressor interactions. This work establishes the BTB domain as a highly druggable structure, paving the way for the use of other members of this protein family as drug targets. The magnitude of effects elicited by this class of BCL6-degrading compounds exceeds that of our equipotent non-degrading inhibitors, suggesting opportunities for the development of BCL6-based lymphoma therapeutics.


Subject(s)
Proteolysis , Proto-Oncogene Proteins c-bcl-6/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Inhibitory Concentration 50 , Kinetics , Models, Molecular , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Domains , Proteolysis/drug effects , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-6/chemistry , Pyrimidines/pharmacology , Structure-Activity Relationship , Ubiquitination/drug effects
19.
J Med Chem ; 59(22): 10147-10162, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27775892

ABSTRACT

Scaffold modification based on Wang's pioneering MDM2-p53 inhibitors led to novel, chemically stable spiro-oxindole compounds bearing a spiro[3H-indole-3,2'-pyrrolidin]-2(1H)-one scaffold that are not prone to epimerization as observed for the initial spiro[3H-indole-3,3'-pyrrolidin]-2(1H)-one scaffold. Further structure-based optimization inspired by natural product architectures led to a complex fused ring system ideally suited to bind to the MDM2 protein and to interrupt its protein-protein interaction (PPI) with TP53. The compounds are highly selective and show in vivo efficacy in a SJSA-1 xenograft model even when given as a single dose as demonstrated for 4-[(3S,3'S,3'aS,5'R,6'aS)-6-chloro-3'-(3-chloro-2-fluorophenyl)-1'-(cyclopropylmethyl)-2-oxo-1,2,3',3'a,4',5',6',6'a-octahydro-1'H-spiro[indole-3,2'-pyrrolo[3,2-b]pyrrole]-5'-yl]benzoic acid (BI-0252).


Subject(s)
Drug Discovery , Indoles/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrrolidinones/pharmacology , Spiro Compounds/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Administration, Oral , Dose-Response Relationship, Drug , Humans , Indoles/administration & dosage , Indoles/chemistry , Models, Molecular , Molecular Structure , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrrolidinones/administration & dosage , Pyrrolidinones/chemistry , Spiro Compounds/administration & dosage , Spiro Compounds/chemistry , Structure-Activity Relationship , Tumor Suppressor Protein p53/metabolism
20.
Nat Chem Biol ; 12(9): 672-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27376689

ABSTRACT

Here we show that acute myeloid leukemia (AML) cells require the BRD9 subunit of the SWI-SNF chromatin-remodeling complex to sustain MYC transcription, rapid cell proliferation and a block in differentiation. Based on these observations, we derived small-molecule inhibitors of the BRD9 bromodomain that selectively suppress the proliferation of mouse and human AML cell lines. To establish these effects as on-target, we engineered a bromodomain-swap allele of BRD9 that retains functionality despite a radically altered bromodomain pocket. Expression of this allele in AML cells confers resistance to the antiproliferative effects of our compound series, thus establishing BRD9 as the relevant cellular target. Furthermore, we used an analogous domain-swap strategy to generate an inhibitor-resistant allele of EZH2. To our knowledge, our study provides the first evidence for a role of BRD9 in cancer and reveals a simple genetic strategy for constructing resistance alleles to demonstrate on-target activity of chemical probes in cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Engineering , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Transcription Factors/antagonists & inhibitors , Alleles , Animals , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL