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










Publication year range
1.
bioRxiv ; 2023 Jan 02.
Article in English | MEDLINE | ID: mdl-36711980

ABSTRACT

While specific cell signaling pathway inhibitors have yielded great success in oncology, directly triggering cancer cell death is one of the great drug discovery challenges facing biomedical research in the era of precision oncology. Attempts to eradicate cancer cells expressing unique target proteins, such as antibody-drug conjugates (ADCs), T-cell engaging therapies, and radiopharmaceuticals have been successful in the clinic, but they are limited by the number of targets given the inability to target intracellular proteins. More recently, heterobifunctional small molecules such as Proteolysis Targeting Chimera (PROTACs) have paved the way for protein proximity inducing therapeutic modalities. Here, we describe a proof-of-concept study using novel heterobifunctional small molecules called Regulated Induced Proximity Targeting Chimeras or RIPTACs, which elicit a stable ternary complex between a target protein selectively expressed in cancer tissue and a pan-expressed protein essential for cell survival. The resulting cooperative protein:protein interaction (PPI) abrogates the function of the essential protein, thus leading to cell death selectively in cells expressing the target protein. This approach not only opens new target space by leveraging differentially expressed intracellular proteins but also has the advantage of not requiring the target to be a driver of disease. Thus, RIPTACs can address non-target mechanisms of resistance given that cell killing is driven by inactivation of the essential protein. Using the HaloTag7-FKBP model system as a target protein, we describe RIPTACs that incorporate a covalent or non-covalent target ligand connected via a linker to effector ligands such as JQ1 (BRD4), BI2536 (PLK1), or multi-CDK inhibitors such as TMX3013 or dinaciclib. We show that these RIPTACs exhibit positive co-operativity, accumulate selectively in cells expressing HaloTag7-FKBP, form stable target:RIPTAC:effector trimers in cells, and induce an anti-proliferative response in target-expressing cells. We propose that RIPTACs are a novel heterobifunctional therapeutic modality to treat cancers that are known to selectively express a specific intracellular protein.

2.
Blood ; 134(1): 59-73, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31023702

ABSTRACT

RUNX1 transcription factor regulates normal and malignant hematopoiesis. Somatic or germline mutant RUNX1 (mtRUNX1) is associated with poorer outcome in acute myeloid leukemia (AML). Knockdown or inhibition of RUNX1 induced more apoptosis of AML expressing mtRUNX1 versus wild-type RUNX1 and improved survival of mice engrafted with mtRUNX1-expressing AML. CRISPR/Cas9-mediated editing-out of RUNX1 enhancer (eR1) within its intragenic super-enhancer, or BET protein BRD4 depletion by short hairpin RNA, repressed RUNX1, inhibited cell growth, and induced cell lethality in AML cells expressing mtRUNX1. Moreover, treatment with BET protein inhibitor or degrader (BET-proteolysis targeting chimera) repressed RUNX1 and its targets, inducing apoptosis and improving survival of mice engrafted with AML expressing mtRUNX1. Library of Integrated Network-based Cellular Signatures 1000-connectivity mapping data sets queried with messenger RNA signature of RUNX1 knockdown identified novel expression-mimickers (EMs), which repressed RUNX1 and exerted in vitro and in vivo efficacy against AML cells expressing mtRUNX1. In addition, the EMs cinobufagin, anisomycin, and narciclasine induced more lethality in hematopoietic progenitor cells (HPCs) expressing germline mtRUNX1 from patients with AML compared with HPCs from patients with familial platelet disorder (FPD), or normal untransformed HPCs. These findings highlight novel therapeutic agents for AML expressing somatic or germline mtRUNX1.


Subject(s)
Antineoplastic Agents/pharmacology , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Core Binding Factor Alpha 2 Subunit/genetics , Gene Knockdown Techniques , Germ-Line Mutation , Hematopoietic Stem Cells/drug effects , Humans , Mice
3.
Leukemia ; 33(6): 1373-1386, 2019 06.
Article in English | MEDLINE | ID: mdl-30575820

ABSTRACT

Transformation of post-myeloproliferative neoplasms into secondary (s) AML exhibit poor clinical outcome. In addition to increased JAK-STAT and PI3K-AKT signaling, post-MPN sAML blast progenitor cells (BPCs) demonstrate increased nuclear ß-catenin levels and TCF7L2 (TCF4) transcriptional activity. Knockdown of ß-catenin or treatment with BC2059 that disrupts binding of ß-catenin to TBL1X (TBL1) depleted nuclear ß-catenin levels. This induced apoptosis of not only JAKi-sensitive but also JAKi-persister/resistant post-MPN sAML BPCs, associated with attenuation of TCF4 transcriptional targets MYC, BCL-2, and Survivin. Co-targeting of ß-catenin and JAK1/2 inhibitor ruxolitinib (rux) synergistically induced lethality in post-MPN sAML BPCs and improved survival of mice engrafted with human sAML BPCs. Notably, co-treatment with BET protein degrader ARV-771 and BC2059 also synergistically induced apoptosis and improved survival of mice engrafted with JAKi-sensitive or JAKi-persister/resistant post-MPN sAML cells. These preclinical findings highlight potentially promising anti-post-MPN sAML activity of the combination of ß-catenin and BETP antagonists against post-MPN sAML BPCs.


Subject(s)
Cell Nucleus/drug effects , Drug Synergism , Leukemia, Myeloid, Acute/drug therapy , Myeloproliferative Disorders/drug therapy , Protein Kinase Inhibitors/pharmacology , beta Catenin/antagonists & inhibitors , Acetanilides/pharmacology , Animals , Apoptosis/drug effects , CRISPR-Cas Systems , Cell Nucleus/metabolism , Cell Nucleus/pathology , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
4.
Leukemia ; 32(10): 2224-2239, 2018 10.
Article in English | MEDLINE | ID: mdl-29581547

ABSTRACT

Bromodomain and extraterminal (BET) domain containing protein (BRD)-4 modulates the expression of oncogenes such as c-myc, and is a promising therapeutic target in diverse cancer types. We performed pre-clinical studies in myeloma models with bi-functional protein-targeting chimeric molecules (PROTACs) which target BRD4 and other BET family members for ubiquitination and proteasomal degradation. PROTACs potently reduced the viability of myeloma cell lines in a time-dependent and concentration-dependent manner associated with G0/G1 arrest, reduced levels of CDKs 4 and 6, increased p21 levels, and induction of apoptosis. These agents specifically decreased cellular levels of downstream BRD4 targets, including c-MYC and N-MYC, and a Cereblon-targeting PROTAC showed downstream effects similar to those of an immunomodulatory agent. Notably, PROTACs overcame bortezomib, dexamethasone, lenalidomide, and pomalidomide resistance, and their activity was maintained in otherwise isogenic myeloma cells with wild-type or deleted TP53. Combination studies showed synergistic interactions with dexamethasone, BH3 mimetics, and Akt pathway inhibitors. BET-specific PROTACs induced a rapid loss of viability of primary cells from myeloma patients, and delayed growth of MM1.S-based xenografts. Our data demonstrate that BET degraders have promising activity against pre-clinical models of multiple myeloma, and support their translation to the clinic for patients with relapsed and/or refractory disease.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Proteins/metabolism , Amino Acid Motifs/drug effects , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mice , Mice, Inbred NOD , Nuclear Proteins/metabolism , Protein Domains/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
5.
J Med Chem ; 61(2): 583-598, 2018 01 25.
Article in English | MEDLINE | ID: mdl-28692295

ABSTRACT

Proteolysis targeting chimeras (PROTACs) are bifunctional molecules that recruit an E3 ligase to a target protein to facilitate ubiquitination and subsequent degradation of that protein. While the field of targeted degraders is still relatively young, the potential for this modality to become a differentiated and therapeutic reality is strong, such that both academic and pharmaceutical institutions are now entering this interesting area of research. In this article, we describe a broadly applicable process for identifying degrader hits based on the serine/threonine kinase TANK-binding kinase 1 (TBK1) and have generalized the key structural elements associated with degradation activities. Compound 3i is a potent hit (TBK1 DC50 = 12 nM, Dmax = 96%) with excellent selectivity against a related kinase IKKε, which was further used as a chemical tool to assess TBK1 as a target in mutant K-Ras cancer cells.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Proteolysis/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Cell Line , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Fluorescence Polarization , Genes, ras , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Molecular Structure , Mutation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Structure-Activity Relationship , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/genetics
6.
Curr Opin Chem Biol ; 39: 46-53, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28605671

ABSTRACT

Small molecule probes of biological systems have traditionally been designed to bind to and inhibit the active sites of their protein targets. While this class of pharmacological agents has been broadened by the development of a small number of allosteric and protein-protein interaction (PPI) inhibitors, conventional drug design still excludes 'undruggable' proteins that are neither enzymes nor receptors. Recent years have seen the emergence of new classes of small molecules that can target hitherto undruggable proteins by recruiting the cellular proteostasis machinery to selectively tag them for degradation. These molecules, especially the class known as Proteolysis Targeting Chimera (PROTACs), represent a paradigm shift in chemical genetics, but their most tantalizing potential is as novel therapeutic agents. This review briefly summarizes the preclinical development of small molecule-based protein degraders, and describes the recent improvements in the technology that have positioned PROTACs on the cusp of entering the clinic.


Subject(s)
Proteins/metabolism , Proteolysis/drug effects , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Animals , Humans , Proteins/chemistry
7.
Proc Natl Acad Sci U S A ; 113(26): 7124-9, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27274052

ABSTRACT

Prostate cancer has the second highest incidence among cancers in men worldwide and is the second leading cause of cancer deaths of men in the United States. Although androgen deprivation can initially lead to remission, the disease often progresses to castration-resistant prostate cancer (CRPC), which is still reliant on androgen receptor (AR) signaling and is associated with a poor prognosis. Some success against CRPC has been achieved by drugs that target AR signaling, but secondary resistance invariably emerges, and new therapies are urgently needed. Recently, inhibitors of bromodomain and extra-terminal (BET) family proteins have shown growth-inhibitory activity in preclinical models of CRPC. Here, we demonstrate that ARV-771, a small-molecule pan-BET degrader based on proteolysis-targeting chimera (PROTAC) technology, demonstrates dramatically improved efficacy in cellular models of CRPC as compared with BET inhibition. Unlike BET inhibitors, ARV-771 results in suppression of both AR signaling and AR levels and leads to tumor regression in a CRPC mouse xenograft model. This study is, to our knowledge, the first to demonstrate efficacy with a small-molecule BET degrader in a solid-tumor malignancy and potentially represents an important therapeutic advance in the treatment of CRPC.


Subject(s)
Antineoplastic Agents/administration & dosage , Nuclear Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle Proteins , Cell Line, Tumor , Humans , Male , Mice , Nuclear Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Protein Serine-Threonine Kinases/genetics , Proteolysis , RNA-Binding Proteins/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics
8.
Chem Biol ; 22(6): 755-63, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26051217

ABSTRACT

BRD4, a bromodomain and extraterminal domain (BET) family member, is an attractive target in multiple pathological settings, particularly cancer. While BRD4 inhibitors have shown some promise in MYC-driven malignancies such as Burkitt's lymphoma (BL), we show that BRD4 inhibitors lead to robust BRD4 protein accumulation, which may account for their limited suppression of MYC expression, modest antiproliferative activity, and lack of apoptotic induction. To address these limitations we designed ARV-825, a hetero-bifunctional PROTAC (Proteolysis Targeting Chimera) that recruits BRD4 to the E3 ubiquitin ligase cereblon, leading to fast, efficient, and prolonged degradation of BRD4 in all BL cell lines tested. Consequently, ARV-825 more effectively suppresses c-MYC levels and downstream signaling than small-molecule BRD4 inhibitors, resulting in more effective cell proliferation inhibition and apoptosis induction in BL. Our findings provide strong evidence that cereblon-based PROTACs provide a better and more efficient strategy in targeting BRD4 than traditional small-molecule inhibitors.


Subject(s)
Azepines/pharmacology , Nuclear Proteins/metabolism , Peptide Hydrolases/metabolism , Signal Transduction/drug effects , Thalidomide/analogs & derivatives , Transcription Factors/metabolism , Acetanilides/toxicity , Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Azepines/chemistry , Azepines/toxicity , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Heterocyclic Compounds, 3-Ring/toxicity , Humans , Nuclear Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Thalidomide/chemistry , Thalidomide/pharmacology , Transcription Factors/antagonists & inhibitors , Triazoles/toxicity , Ubiquitin-Protein Ligases
9.
ACS Chem Biol ; 10(8): 1831-7, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26070106

ABSTRACT

Small molecule-induced protein degradation is an attractive strategy for the development of chemical probes. One method for inducing targeted protein degradation involves the use of PROTACs, heterobifunctional molecules that can recruit specific E3 ligases to a desired protein of interest. PROTACs have been successfully used to degrade numerous proteins in cells, but the peptidic E3 ligase ligands used in previous PROTACs have hindered their development into more mature chemical probes or therapeutics. We report the design of a novel class of PROTACs that incorporate small molecule VHL ligands to successfully degrade HaloTag7 fusion proteins. These HaloPROTACs will inspire the development of future PROTACs with more drug-like properties. Additionally, these HaloPROTACs are useful chemical genetic tools, due to their ability to chemically knock down widely used HaloTag7 fusion proteins in a general fashion.


Subject(s)
Proteolysis , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Ubiquitin-Protein Ligases/metabolism , HEK293 Cells , Humans , Ligands , Protein Binding
10.
Nat Chem Biol ; 10(11): 957-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25242550

ABSTRACT

Accumulation of unfolded proteins within the endoplasmic reticulum (ER) of eukaryotic cells leads to an unfolded protein response (UPR) that either restores homeostasis or commits the cells to apoptosis. Tools traditionally used to study the UPR are proapoptotic and thus confound analysis of long-term cellular responses to ER stress. Here, we describe an ER-localized HaloTag (ERHT) protein that can be conditionally destabilized using a small-molecule hydrophobic tag (HyT36). Treatment of ERHT-expressing cells with HyT36 induces acute, resolvable ER stress that results in transient UPR activation without induction of apoptosis. Transcriptome analysis of late-stage responses to this UPR stimulus reveals a link between UPR activity and estrogen signaling.


Subject(s)
Adamantane/analogs & derivatives , Endoplasmic Reticulum Stress/drug effects , Estrogens/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Stability/drug effects , Unfolded Protein Response/drug effects , Adamantane/chemistry , Adamantane/pharmacology , Apoptosis , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Signal Transduction/drug effects , Thapsigargin/pharmacology , Tunicamycin/pharmacology , Up-Regulation/drug effects
11.
Chembiochem ; 13(4): 538-41, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22271667

ABSTRACT

New HyTs are a knockout: we previously reported that labeling HaloTag proteins with low molecular weight hydrophobic tags (HyTs) leads to targeted degradation of HaloTag fusion proteins. In this report, we employed a chemical approach to extend this hydrophobic tagging methodology to highly stabilized proteins by synthesizing and evaluating a library of HyTs, which led to the identification of HyT36.


Subject(s)
Recombinant Fusion Proteins/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Structure , Recombinant Fusion Proteins/chemistry
12.
Nat Chem Biol ; 7(8): 538-43, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725302

ABSTRACT

The ability to regulate any protein of interest in living systems with small molecules remains a challenge. We hypothesized that appending a hydrophobic moiety to the surface of a protein would mimic the partially denatured state of the protein, thus engaging the cellular quality control machinery to induce its proteasomal degradation. We designed and synthesized bifunctional small molecules to bind a bacterial dehalogenase (the HaloTag protein) and present a hydrophobic group on its surface. Hydrophobic tagging of the HaloTag protein with an adamantyl moiety induced the degradation of cytosolic, isoprenylated and transmembrane HaloTag fusion proteins in cell culture. We demonstrated the in vivo utility of hydrophobic tagging by degrading proteins expressed in zebrafish embryos and by inhibiting Hras1(G12V)-driven tumor progression in mice. Therefore, hydrophobic tagging of HaloTag fusion proteins affords small-molecule control over any protein of interest, making it an ideal system for validating potential drug targets in disease models.


Subject(s)
Biosensing Techniques/methods , Fluorescent Dyes/chemistry , Animals , Cell Line , Humans , Hydrophobic and Hydrophilic Interactions , Luminescent Proteins/chemistry , Mice , Molecular Structure , Recombinant Proteins , Sensitivity and Specificity , Staining and Labeling , Zebrafish
13.
J Biol Chem ; 285(15): 11057-60, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20147751

ABSTRACT

The functions of many cellular proteins have been elucidated by selective gene inactivation and subsequent phenotypic analysis. For example, genetic mutations, gene knock-out generation, and the use of RNA interference to target mRNA for degradation can all result in decreased production of a specific protein, yielding informative cellular phenotypes. However, these techniques each have certain inherent limitations. This minireview focuses on the recent development of new approaches to study protein function at the post-translational level, namely chemical induction of targeted protein degradation.


Subject(s)
Biochemistry/methods , Proteins/chemistry , RNA Interference , Animals , Humans , Models, Biological , Peptide Hydrolases/chemistry , Phenotype , Proteasome Endopeptidase Complex/chemistry , Protein Processing, Post-Translational , Protein Structure, Tertiary , RNA, Messenger/metabolism , Ubiquitin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...