Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Cell ; 181(1): 102-114, 2020 04 02.
Article in English | MEDLINE | ID: mdl-31955850

ABSTRACT

New biological tools provide new techniques to probe fundamental biological processes. Here we describe the burgeoning field of proteolysis-targeting chimeras (PROTACs), which are capable of modulating protein concentrations at a post-translational level by co-opting the ubiquitin-proteasome system. We describe the PROTAC technology and its application to drug discovery and provide examples where PROTACs have enabled novel biological insights. Furthermore, we provide a workflow for PROTAC development and use and discuss the benefits and issues associated with PROTACs. Finally, we compare PROTAC-mediated protein-level modulation with other technologies, such as RNAi and genome editing.


Subject(s)
Drug Discovery , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Humans
2.
Angew Chem Int Ed Engl ; 63(14): e202316496, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38348945

ABSTRACT

Brachyury is an oncogenic transcription factor whose overexpression drives chordoma growth. The downmodulation of brachyury in chordoma cells has demonstrated therapeutic potential, however, as a transcription factor it is classically deemed "undruggable". Given that direct pharmacological intervention against brachyury has proven difficult, attempts at intervention have instead targeted upstream kinases. Recently, afatinib, an FDA-approved kinase inhibitor, has been shown to modulate brachyury levels in multiple chordoma cell lines. Herein, we use afatinib as a lead to undertake a structure-based drug design approach, aided by mass-spectrometry and X-ray crystallography, to develop DHC-156, a small molecule that more selectively binds brachyury and downmodulates it as potently as afatinib. We eliminated kinase-inhibition from this novel scaffold while demonstrating that DHC-156 induces the post-translational downmodulation of brachyury that results in an irreversible impairment of chordoma tumor cell growth. In doing so, we demonstrate the feasibility of direct brachyury modulation, which may further be developed into more potent tool compounds and therapies.


Subject(s)
Chordoma , Fetal Proteins , Transcription Factors , Humans , Transcription Factors/metabolism , Chordoma/drug therapy , Chordoma/metabolism , Chordoma/pathology , Afatinib , T-Box Domain Proteins/metabolism
3.
J Am Chem Soc ; 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36753634

ABSTRACT

Microtubule-associated protein tau is essential for microtubule assembly and stabilization. Hyperphosphorylation of the microtubule-associated protein tau plays an important pathological role in the development of Alzheimer's disease and other tauopathies. In vivo studies using kinase inhibitors suggest that reducing tau phosphorylation levels has therapeutic potential; however, such approaches showed limited benefits. We sought to further develop our phosphorylation targeting chimera (PhosTAC) technology to specifically induce tau dephosphorylation. Herein, we use small molecule-based PhosTACs to recruit tau to PP2A, a native tau phosphatase. PhosTACs induced the formation of a stable ternary complex, leading to rapid, efficient, and sustained tau dephosphorylation, which also correlated with the enhanced downregulation of tau protein. Mass spectrometry data validated that PhosTACs downregulated multiple phosphorylation sites of tau. We believe that PhosTAC possesses several advantages over current strategies to modulate tau phosphorylation and represents a new avenue for disease-modifying therapies for tauopathies.

4.
Chem Soc Rev ; 51(12): 5214-5236, 2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35671157

ABSTRACT

Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules consisting of one ligand that binds to a protein of interest (POI) and another that can recruit an E3 ubiquitin ligase. The chemically-induced proximity between the POI and E3 ligase results in ubiquitination and subsequent degradation of the POI by the ubiquitin-proteasome system (UPS). The event-driven mechanism of action (MOA) of PROTACs offers several advantages compared to traditional occupancy-driven small molecule inhibitors, such as a catalytic nature, reduced dosing and dosing frequency, a more potent and longer-lasting effect, an added layer of selectivity to reduce potential toxicity, efficacy in the face of drug-resistance mechanisms, targeting nonenzymatic functions, and expanded target space. Here, we highlight important milestones and briefly discuss lessons learned about targeted protein degradation (TPD) in recent years and conjecture on the efforts still needed to expand the toolbox for PROTAC discovery to ultimately provide promising therapeutics.


Subject(s)
Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
J Biol Chem ; 296: 100647, 2021.
Article in English | MEDLINE | ID: mdl-33839157

ABSTRACT

Of late, targeted protein degradation (TPD) has surfaced as a novel and innovative chemical tool and therapeutic modality. By co-opting protein degradation pathways, TPD facilitates complete removal of the protein molecules from within or outside the cell. While the pioneering Proteolysis-Targeting Chimera (PROTAC) technology and molecular glues hijack the ubiquitin-proteasome system, newer modalities co-opt autophagy or the endo-lysosomal pathway. Using this mechanism, TPD is posited to largely expand the druggable space far beyond small-molecule inhibitors. In this review, we discuss the major advances in TPD, highlight our current understanding, and explore outstanding questions in the field.


Subject(s)
Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Proteolysis , Ubiquitination
6.
J Am Chem Soc ; 144(12): 5594-5605, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35311258

ABSTRACT

Targeted protein degradation (TPD) by PROTACs is a promising strategy to control disease-causing protein levels within the cell. While TPD is emerging as an innovative drug discovery paradigm, there are currently only a limited number of E3 ligase:ligand pairs that are employed to induce protein degradation. Herein, we report a novel approach to induce protein degradation by hijacking a methyl reader:E3 ligase complex. L3MBTL3 is a methyl-lysine reader protein that binds to the Cul4DCAF5 E3 ligase complex and targets methylated proteins for proteasomal degradation. By co-opting this natural mechanism, we report the design and biological evaluation of L3MBTL3-recruiting PROTACs and demonstrate nuclear-specific degradation of FKBP12 and BRD2. We envision this as a generalizable approach to utilize other reader protein-associated E3 ligase complexes in PROTAC design to expand the E3 ligase toolbox and explore the full potential of TPD.


Subject(s)
Nuclear Proteins , Ubiquitin-Protein Ligases , Drug Discovery , Ligands , Nuclear Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism
7.
Chembiochem ; 23(2): e202100270, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34494353

ABSTRACT

Proteolysis-targeting chimeras (PROTACs), an emerging paradigm-shifting technology, hijacks the ubiquitin-proteasome system for targeted protein degradation. PROTACs induce ternary complexes between an E3 ligase and POI, and this induced proximity leads to polyUb chain formation on substrates and eventual proteasomal-mediated POI degradation. PROTACs have shown great therapeutic potential by degrading many disease-causing proteins, such as the androgen receptor and BRD4. The PROTAC technology has advanced significantly in the last two decades, with the repertoire of PROTAC targets increased tremendously. Herein, we describe recent developments of PROTAC technology, focusing on mechanistic and kinetic studies, pharmacokinetic study, spatiotemporal control of PROTACs, covalent PROTACs, resistance to PROTACs, and new E3 ligands.


Subject(s)
Proteins/metabolism , HeLa Cells , Humans , Immunoconjugates/metabolism , Kinetics , Ligands , Light , Protein Conformation , Proteins/chemistry , Proteolysis , Transcription Factors/metabolism
8.
Blood ; 135(15): 1255-1269, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32068780

ABSTRACT

The promising activity of BET protein inhibitors (BETi's) is compromised by adaptive or innate resistance in acute myeloid leukemia (AML). Here, modeling of BETi-persister/resistance (BETi-P/R) in human postmyeloproliferative neoplasm (post-MPN) secondary AML (sAML) cells demonstrated accessible and active chromatin in specific superenhancers/enhancers, which was associated with increased levels of nuclear ß-catenin, TCF7L2, JMJD6, and c-Myc in BETi-P/R sAML cells. Following BETi treatment, c-Myc levels were rapidly restored in BETi-P/R sAML cells. CRISPR/Cas9-mediated knockout of TCF7L2 or JMJD6 reversed BETi-P/R, whereas ectopic overexpression conferred BETi-P/R in sAML cells, confirming the mechanistic role of the ß-catenin-TCF7L2-JMJD6-c-Myc axis in BETi resistance. Patient-derived, post-MPN, CD34+ sAML blasts exhibiting relative resistance to BETi, as compared with sensitive sAML blasts, displayed higher messenger RNA and protein expression of TCF7L2, JMJD6, and c-Myc and following BETi washout exhibited rapid restoration of c-Myc and JMJD6. CRISPR/Cas9 knockout of TCF7L2 and JMJD6 depleted their levels, inducing loss of viability of the sAML blasts. Disruption of colocalization of nuclear ß-catenin with TBL1 and TCF7L2 by the small-molecule inhibitor BC2059 combined with depletion of BRD4 by BET proteolysis-targeting chimera reduced c-Myc levels and exerted synergistic lethality in BETi-P/R sAML cells. This combination also reduced leukemia burden and improved survival of mice engrafted with BETi-P/R sAML cells or patient-derived AML blasts innately resistant to BETi. Therefore, multitargeted disruption of the ß-catenin-TCF7L2-JMJD6-c-Myc axis overcomes adaptive and innate BETi resistance, exhibiting preclinical efficacy against human post-MPN sAML cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Myeloid, Acute/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism
9.
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
10.
J Org Chem ; 86(12): 8479-8488, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34047555

ABSTRACT

A novel, facile, and expeditious two-step synthesis of 3,4-unsubstituted isoquinolin-1(2H)-ones from a Suzuki cross-coupling between 2-halobenzonitriles and commercially available vinyl boronates followed by platinum-catalyzed nitrile hydrolysis and cyclization is described.


Subject(s)
Nitriles , Catalysis , Cyclization
11.
Dev Dyn ; 249(4): 465-482, 2020 04.
Article in English | MEDLINE | ID: mdl-32124513

ABSTRACT

Fifty years ago, Lewis Wolpert defined an important question in developmental biology: how are cell fates determined by the positions of cells within a system? He proposed that cells retain positional values as if they lie within a coordinate system and that the interpretation of these values produces patterns in development. He referred to this concept as positional information. Though initially controversial, this concept of positional information has proven to be profoundly influential in developmental biology. One area in which the influence of Wolpert's theoretical work can be clearly demonstrated is the study of limb regeneration in salamanders. Here, we review the work in limb regeneration leading up to Wolpert defining the concept of positional information and how his theory has guided regeneration research over the subsequent 50 years.


Subject(s)
Extremities/physiology , Regeneration/physiology , Urodela/physiology , Animals , Developmental Biology/methods , Models, Theoretical
12.
Annu Rev Pharmacol Toxicol ; 57: 107-123, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27732798

ABSTRACT

Protein homeostasis networks are highly regulated systems responsible for maintaining the health and productivity of cells. Whereas therapeutics have been developed to disrupt protein homeostasis, more recently identified techniques have been used to repurpose homeostatic networks to effect degradation of disease-relevant proteins. Here, we review recent advances in the use of small molecules to degrade proteins in a selective manner. First, we highlight all-small-molecule techniques with direct clinical application. Second, we describe techniques that may find broader acceptance in the biomedical research community that require little or no synthetic chemistry. In addition to serving as innovative research tools, these new approaches to control intracellular protein levels offer the potential to develop novel therapeutics targeting proteins that are not currently pharmaceutically vulnerable.


Subject(s)
Drug Delivery Systems/methods , Proteins/metabolism , Proteolysis/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/metabolism , Animals , Drug Delivery Systems/trends , Humans , Oligopeptides/administration & dosage , Oligopeptides/metabolism , Proteins/antagonists & inhibitors
13.
Bioorg Med Chem Lett ; 30(3): 126877, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31879210

ABSTRACT

A new series of Proteolysis Targeting Chimeras (PROTACs) targeting Bruton's Tyrosine Kinase (BTK) was synthesized, with the goal of improving the pharmacokinetic properties of our previously reported PROTAC, MT802. We recently described the ability of MT802 to induce degradation of both wild-type and C481S mutant BTK in immortalized cells and patient-derived B-lymphocytes. However, the pharmacokinetic properties of MT802 were not suitable for further in vivo development. Therefore, we undertook a systematic medicinal chemistry campaign to overcome this issue and made a series of PROTACs with structural modifications to the linker and E3-recruiting ligand; more specifically, the new PROTACs were synthesized with different von Hippel-Lindau (VHL) and cereblon (CRBN) ligands while keeping the BTK ligand and linker length constant. This approach resulted in an equally potent PROTAC, SJF620, with a significantly better pharmacokinetic profile than MT802. This compound may hold promise for further in vivo exploration of BTK degradation.


Subject(s)
Acetamides/chemistry , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Drug Design , Ligands , Acetamides/chemical synthesis , Acetamides/pharmacokinetics , Acetamides/pharmacology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/metabolism , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Binding Sites , Cell Line , Half-Life , Humans , Molecular Docking Simulation , Mutagenesis, Site-Directed , Piperidines/chemistry , Piperidines/metabolism , Protein Structure, Tertiary , Proteolysis/drug effects , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
14.
Chem Rev ; 117(17): 11269-11301, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28777566

ABSTRACT

Control of protein levels by nucleic-acid-based technologies has proven to be a useful research tool but lacks the advantages of small molecules with respect to cell permeability, temporal control, and the potential generation of therapeutics. In this Review, we discuss the technologies available for the control of intracellular protein levels with small molecules and compare the various systems available.


Subject(s)
Proteins/metabolism , Small Molecule Libraries/chemistry , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Small Molecule Libraries/metabolism , Ubiquitin/antagonists & inhibitors , Ubiquitin/metabolism
15.
Nature ; 554(7690): 34-35, 2018 Feb.
Article in English | MEDLINE | ID: mdl-32080442
16.
Nature ; 554(7690): 34-35, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29388980
17.
Drug Discov Today Technol ; 31: 15-27, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31200855

ABSTRACT

The majority of currently used therapeutics are small molecule-based and utilize occupancy-driven pharmacology as the mode of action (MOA), in which the protein function is modulated via temporary inhibition. New modalities that operate using alternative MOAs are essential for tapping into the "undruggable" proteome. The PROteolysis Targeting Chimera (PROTAC) technology provides an attractive new approach that utilizes an event-driven MOA. Small molecule-based heterobifunctional PROTACs modulate protein target levels by hijacking the ubiquitin-proteasome system to induce degradation of the target. Here, we address important milestones in the development of the PROTAC technology, as well as emphasize key findings from this previous year and highlight future directions of this promising drug discovery modality.


Subject(s)
Drug Discovery , Proteolysis , Animals , Humans , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
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
19.
Biochemistry ; 57(26): 3564-3575, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29851337

ABSTRACT

Inhibition of Bruton's tyrosine kinase (BTK) with the irreversible inhibitor ibrutinib has emerged as a transformative treatment option for patients with chronic lymphocytic leukemia (CLL) and other B-cell malignancies, yet >80% of CLL patients develop resistance due to a cysteine to serine mutation at the site covalently bound by ibrutinib (C481S). Currently, an effective treatment option for C481S patients exhibiting relapse to ibrutinib does not exist, and these patients have poor outcomes. To address this, we have developed a PROteolysis TArgeting Chimera (PROTAC) that induces degradation of both wild-type and C481S mutant BTK. We selected a lead PROTAC, MT-802, from several candidates on the basis of its potency to induce BTK knockdown. MT-802 recruits BTK to the cereblon E3 ubiquitin ligase complex to trigger BTK ubiquitination and degradation via the proteasome. MT-802 binds fewer off-target kinases than ibrutinib does and retains an equivalent potency (>99% degradation at nanomolar concentrations) against wild-type and C481S BTK. In cells isolated from CLL patients with the C481S mutation, MT-802 is able to reduce the pool of active, phosphorylated BTK whereas ibrutinib cannot. Collectively, these data provide a basis for further preclinical study of BTK PROTACs as a novel strategy for treatment of C481S mutant CLL.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/genetics , Amino Acid Substitution , Drug Resistance, Neoplasm , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/metabolism , Cell Line, Tumor , Drug Design , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Molecular Docking Simulation , Piperidines , Point Mutation , Protein Kinase Inhibitors/chemistry , Proteolysis/drug effects , Pyrazoles/chemistry , Pyrimidines/chemistry , Ubiquitination/drug effects
20.
J Am Chem Soc ; 140(49): 17019-17026, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30444612

ABSTRACT

Enzymatic inhibition has proven to be a successful modality for the development of many small-molecule drugs. In recent years, small-molecule-induced protein degradation has emerged as an orthogonal therapeutic strategy that has the potential to expand the druggable target space. Focal adhesion kinase (Fak) is a key player in tumor invasion and metastasis, acting simultaneously as a kinase and a scaffold for several signaling proteins. While previous efforts to modulate Fak activity were limited to kinase inhibitors with low success in clinical studies, protein degradation offers a possibility to simultaneously block Fak's kinase signaling and scaffolding capabilities. Here, we report the development of a selective and potent Fak degrader, PROTAC-3, which outperforms a clinical candidate, defactinib, with respect to Fak activation as well as Fak-mediated cell migration and invasion. These results underline the potential that PROTACs offer in expanding the druggable space and controlling protein functions that are not easily addressed by traditional small-molecule therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Focal Adhesion Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Sulfonamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzamides/chemical synthesis , Benzamides/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Drug Design , Focal Adhesion Kinase 1/metabolism , Humans , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proteolysis/drug effects , Pyrazines/chemical synthesis , Pyrazines/chemistry , Signal Transduction/drug effects , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL