Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
J Med Chem ; 65(19): 13052-13073, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36178776

ABSTRACT

Addressing resistance to third-generation EGFR TKIs such as osimertinib via the EGFRC797S mutation remains a highly unmet need in EGFR-driven non-small-cell lung cancer (NSCLC). Herein, we present the discovery of the allosteric EGFR inhibitor 57, a novel fourth-generation inhibitor to overcome EGFRC797S-mediated resistance in patients harboring the activating EGFRL858R mutation. 57 exhibits an improved potency compared to previous allosteric EGFR inhibitors. To our knowledge, 57 is the first allosteric EGFR inhibitor that demonstrates robust tumor regression in a mutant EGFRL858R/C797S tumor model. Additionally, 57 is active in an H1975 EGFRL858R/T790M NSCLC xenograft model and shows superior efficacy in combination with osimertinib compared to the single agents. Our data highlight the potential of 57 as a single agent against EGFRL858R/C797S and EGFRL858R/T790M/C797S and as combination therapy for EGFRL858R- and EGFRL858R/T790M-driven NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Acrylamides , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Humans , Indoles , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines
2.
Clin Cancer Res ; 28(4): 770-780, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34782366

ABSTRACT

PURPOSE: Disease progression in BRAF V600E/K positive melanomas to approved BRAF/MEK inhibitor therapies is associated with the development of resistance mediated by RAF dimer inducing mechanisms. Moreover, progressing disease after BRAFi/MEKi frequently involves brain metastasis. Here we present the development of a novel BRAF inhibitor (Compound Ia) designed to address the limitations of available BRAFi/MEKi. EXPERIMENTAL DESIGN: The novel, brain penetrant, paradox breaker BRAFi is comprehensively characterized in vitro, ex vivo, and in several preclinical in vivo models of melanoma mimicking peripheral disease, brain metastatic disease, and acquired resistance to first-generation BRAFi. RESULTS: Compound Ia manifested elevated potency and selectivity, which triggered cytotoxic activity restricted to BRAF-mutated models and did not induce RAF paradoxical activation. In comparison to approved BRAFi at clinical relevant doses, this novel agent showed a substantially improved activity in a number of diverse BRAF V600E models. In addition, as a single agent, it outperformed a currently approved BRAFi/MEKi combination in a model of acquired resistance to clinically available BRAFi. Compound Ia presents high central nervous system (CNS) penetration and triggered evident superiority over approved BRAFi in a macro-metastatic and in a disseminated micro-metastatic brain model. Potent inhibition of MAPK by Compound Ia was also demonstrated in patient-derived tumor samples. CONCLUSIONS: The novel BRAFi demonstrates preclinically the potential to outperform available targeted therapies for the treatment of BRAF-mutant tumors, thus supporting its clinical investigation.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Brain/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
3.
ACS Chem Biol ; 14(10): 2215-2223, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31553577

ABSTRACT

Proteolysis targeting chimeras are bifunctional small molecules capable of recruiting a target protein of interest to an E3 ubiquitin ligase that facilitates target ubiquitination followed by proteasome-mediated degradation. The first molecules acting on this novel therapeutic paradigm have just entered clinical testing. Here, by using Bromodomain Containing 4 (BRD4) degraders engaging cereblon and Von Hippel-Lindau E3 ligases, we investigated key determinants of resistance to this new mode of action. A loss-of-function screen for genes required for BRD4 degradation revealed strong dependence on the E2 and E3 ubiquitin ligases as well as for members of the COP9 signalosome complex for both cereblon- and Von Hippel-Lindau-engaging BRD4 degraders. Cancer cell lines raised to resist BRD4 degraders manifested a degrader-specific mechanism of resistance, resulting from the loss of components of the ubiquitin proteasome system. In addition, degrader profiling in a cancer cell line panel revealed a differential pattern of activity of Von Hippel-Lindau- and cereblon-based degraders, highlighting the need for the identification of degradation-predictive biomarkers enabling effective patient stratification.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Azepines/pharmacology , Cell Cycle Proteins/metabolism , Drug Resistance/drug effects , Transcription Factors/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Cell Cycle Proteins/chemistry , Cell Line, Tumor , Dipeptides/pharmacology , HEK293 Cells , Humans , Phthalimides/pharmacology , Proof of Concept Study , Proteolysis , Transcription Factors/chemistry , Ubiquitin-Protein Ligases/metabolism
4.
Angew Chem Int Ed Engl ; 56(5): 1294-1297, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27981705

ABSTRACT

CSN5 is the zinc metalloprotease subunit of the COP9 signalosome (CSN), which is an important regulator of cullin-RING E3 ubiquitin ligases (CRLs). CSN5 is responsible for the cleavage of NEDD8 from CRLs, and blocking deconjugation of NEDD8 traps the CRLs in a hyperactive state, thereby leading to auto-ubiquitination and ultimately degradation of the substrate recognition subunits. Herein, we describe the discovery of azaindoles as a new class of CSN5 inhibitors, which interact with the active-site zinc ion of CSN5 through an unprecedented binding mode. The best compounds inhibited CSN5 with nanomolar potency, led to degradation of the substrate recognition subunit Skp2 in cells, and reduced the viability of HCT116 cells.


Subject(s)
COP9 Signalosome Complex/antagonists & inhibitors , Indoles/metabolism , Zinc/metabolism , Binding Sites , COP9 Signalosome Complex/genetics , COP9 Signalosome Complex/metabolism , Catalytic Domain , Cell Proliferation/drug effects , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , HCT116 Cells , Humans , Indoles/chemistry , Indoles/pharmacology , Molecular Docking Simulation , NEDD8 Protein/chemistry , NEDD8 Protein/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , RNA, Small Interfering/metabolism , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/metabolism , Zinc/chemistry
5.
Nat Commun ; 7: 13166, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27774986

ABSTRACT

The COP9 signalosome (CSN) is a central component of the activation and remodelling cycle of cullin-RING E3 ubiquitin ligases (CRLs), the largest enzyme family of the ubiquitin-proteasome system in humans. CRLs are implicated in the regulation of numerous cellular processes, including cell cycle progression and apoptosis, and aberrant CRL activity is frequently associated with cancer. Remodelling of CRLs is initiated by CSN-catalysed cleavage of the ubiquitin-like activator NEDD8 from CRLs. Here we describe CSN5i-3, a potent, selective and orally available inhibitor of CSN5, the proteolytic subunit of CSN. The compound traps CRLs in the neddylated state, which leads to inactivation of a subset of CRLs by inducing degradation of their substrate recognition module. CSN5i-3 differentially affects the viability of tumour cell lines and suppresses growth of a human xenograft in mice. Our results provide insights into how CSN regulates CRLs and suggest that CSN5 inhibition has potential for anti-tumour therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , COP9 Signalosome Complex/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lymphoma, Large-Cell, Anaplastic/drug therapy , Pyrazoles/pharmacology , Ubiquitin-Protein Ligases/genetics , Animals , Antineoplastic Agents/chemical synthesis , Azepines/chemical synthesis , COP9 Signalosome Complex/genetics , COP9 Signalosome Complex/metabolism , Female , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Mice , Mice, SCID , Molecular Targeted Therapy , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Protein Processing, Post-Translational , Proteolysis/drug effects , Pyrazoles/chemical synthesis , THP-1 Cells , Tumor Burden/drug effects , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
6.
J Exp Med ; 210(1): 23-30, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23267013

ABSTRACT

B cell development requires tight regulation to allow for the generation of a diverse repertoire while preventing the development of autoreactive cells. We report, using N-ethyl-N-nitrosourea (ENU)-induced mutagenesis, the identification of a mutant mouse (chompB) with a block in early B cell development. The blockade occurs after the transitional 1 (T1) stage and leads to a decrease in mature B cell subsets and deficits in T cell-dependent antibody responses. Additionally, chompB mice have decreases in myeloid dendritic cells (DCs). The mutation was mapped to the intramembrane protease signal peptide peptidase-like 2a (Sppl2a), a gene not previously implicated in immune cell development. Proteomic analysis identified the invariant chain (CD74) as a key substrate of Sppl2a and suggests that regulated intramembrane proteolysis of CD74 by Sppl2a contributes to B cell and DC survival. Moreover, these data suggest that modulation of Sppl2a may be a useful therapeutic strategy for treatment of B cell dependent autoimmune disorders.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , B-Lymphocytes/physiology , Dendritic Cells/pathology , Membrane Proteins/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Aspartic Acid Endopeptidases/genetics , B-Lymphocytes/pathology , Cell Survival , Dendritic Cells/physiology , Ethylnitrosourea/pharmacology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Immunoglobulins/metabolism , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mutagenesis/drug effects , Mutation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
J Immunol ; 188(6): 2794-804, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22345649

ABSTRACT

Human CMV (HCMV)-encoded NK cell-evasion functions include an MHC class I homolog (UL18) with high affinity for the leukocyte inhibitory receptor-1 (CD85j, ILT2, or LILRB1) and a signal peptide (SP(UL40)) that acts by upregulating cell surface expression of HLA-E. Detailed characterization of SP(UL40) revealed that the N-terminal 14 aa residues bestowed TAP-independent upregulation of HLA-E, whereas C region sequences delayed processing of SP(UL40) by a signal peptide peptidase-type intramembrane protease. Most significantly, the consensus HLA-E-binding epitope within SP(UL40) was shown to promote cell surface expression of both HLA-E and gpUL18. UL40 was found to possess two transcription start sites, with utilization of the downstream site resulting in translation being initiated within the HLA-E-binding epitope (P2). Remarkably, this truncated SP(UL40) was functional and retained the capacity to upregulate gpUL18 but not HLA-E. Thus, our findings identify an elegant mechanism by which an HCMV signal peptide differentially regulates two distinct NK cell-evasion pathways. Moreover, we describe a natural SP(UL40) mutant that provides a clear example of an HCMV clinical virus with a defect in an NK cell-evasion function and exemplifies issues that confront the virus when adapting to immunogenetic diversity in the host.


Subject(s)
Capsid Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Immune Evasion/immunology , Killer Cells, Natural/immunology , Viral Proteins/metabolism , Amino Acid Sequence , Blotting, Northern , Blotting, Western , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Separation , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus/metabolism , Cytomegalovirus Infections , Flow Cytometry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/metabolism , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/genetics , Viral Proteins/immunology , HLA-E Antigens
8.
J Biol Chem ; 283(15): 9966-76, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18270201

ABSTRACT

N-terminal signal sequences mediate endoplasmic reticulum (ER) targeting and insertion of nascent secretory and membrane proteins and are, in most cases, cleaved off by signal peptidase. The mouse mammary tumor virus envelope protein and its alternative splice variant Rem have an unusually long signal sequence, which contains a nuclear localization signal. Although the envelope protein is targeted to the ER, inserted, and glycosylated, Rem has been described as a nuclear protein. Rem as well as a truncated version identical to the cleaved signal sequence have been shown to function as nuclear export factors for intron-containing transcripts. Using transiently transfected cells, we found that Rem is targeted to the ER, where the C-terminal portion is translocated and glycosylated. The signal sequence is cleaved off and accumulates in nucleoli. In a cell-free in vitro system, the generation of the Rem signal peptide depends on the presence of microsomal membranes. In vitro and in cells, the signal peptide initially accumulates in the membrane and is subsequently released into the cytosol. This release does not depend on processing by signal peptide peptidase, an intramembrane cleaving protease that can mediate the liberation of signal peptide fragments from the ER membrane. Our study suggests a novel pathway by which a signal peptide can be released from the ER membrane to fulfill a post-targeting function in a different compartment.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Mammary Tumor Virus, Mouse/metabolism , Nuclear Localization Signals/metabolism , Viral Envelope Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , COS Cells , Chlorocebus aethiops , Cytoplasm/genetics , Cytoplasm/metabolism , Endoplasmic Reticulum/genetics , Glycosylation , HeLa Cells , Humans , Mammary Tumor Virus, Mouse/genetics , Mice , Microsomes/metabolism , Nuclear Localization Signals/genetics , Protein Modification, Translational/physiology , Protein Structure, Tertiary/physiology , Viral Envelope Proteins/genetics
9.
Anal Biochem ; 371(2): 201-7, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17869210

ABSTRACT

The dynamic modification of proteins with ubiquitin is a key regulation paradigm in eukaryotic cells that controls stability, localization, and function of the vast majority of intracellular proteins. Here we describe a robust fluorescence intensity assay for monitoring the enzymatic activity of deubiquitinating proteases, which reverse ubiquitin modifications and comprise over 100 members in humans. The assay was developed for the catalytic domain of human ubiquitin-specific protease 2 (USP2) and human ubiquitin carboxyterminal hydrolase L3 (UCH-L3), and makes use of the novel substrate ubiquitin-rhodamine110-glycine. The latter combines the advantages of a high dynamic range and beneficial optical properties. Its enzymatic behavior is characterized by the kinetic constants K(m)=1.5 microM, k(cat) = 0.53s(-1) and k(cat)/K(m) = 3.5 x 10(5)M(-1) s(-1) for USP2 and K(m) = 34 nM, k(cat)=4.72s(-1), and k(cat)/K(m) = 1.4 x 10(8)M(-1) s(-1) for UCH-L3. This new assay is suitable for inhibitor screening and characterizations, and has been established for the 384-well plate format using protease concentrations of 120 pM for USP2 and 1 pM for UCH-L3 and substrate concentrations of 100 nM for both enzymes. Due to the low protease concentrations and high sensitivity, this assay would allow the determination of inhibitory constants in the subnanomolar range.


Subject(s)
Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Fluorescent Dyes/chemistry , Glycine/analogs & derivatives , Glycine/chemistry , Ubiquitin/chemistry , Biological Assay/instrumentation , Biological Assay/methods , Cysteine Endopeptidases/chemistry , Endopeptidases/chemistry , Fluorescent Dyes/metabolism , Glycine/metabolism , Humans , Inteins , Kinetics , Models, Biological , Rhodamines/chemistry , Rhodamines/metabolism , Spectrometry, Fluorescence , Substrate Specificity , Ubiquitin/metabolism , Ubiquitin Thiolesterase
10.
Curr Pharm Des ; 13(3): 271-85, 2007.
Article in English | MEDLINE | ID: mdl-17313361

ABSTRACT

Aspartic proteases are the smallest class of human proteases with only 15 members. Over the past years, they have received considerable attention as potential targets for pharmaceutical intervention since many have been shown to play important roles in physiological and pathological processes. Despite numerous efforts, however, the only inhibitors for aspartic proteases currently on the market are directed against the HIV protease, an aspartic protease of viral origin. Nevertheless, several inhibitors including those targeting renin, BACE1 and gamma-secretase are in clinical or preclinical development, and some other aspartic proteases are discussed as potential drug target. The crystal structures of seven human aspartic proteases have now been solved and, together with a detailed kinetic understanding of their catalytic mechanism, this has greatly contributed to the design and discovery of novel inhibitors for this protease class. This review describes current aspartic protease drug targets and summarizes the drug discovery efforts in this field. In addition, it highlights recent developments which may lead to a new generation of aspartic protease inhibitors.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/chemistry , Drug Design , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/chemistry , Animals , Aspartic Acid Endopeptidases/metabolism , Computer-Aided Design , HIV Protease/chemistry , HIV Protease Inhibitors/pharmacology , Humans , Membrane Proteins/metabolism , Models, Molecular , Molecular Structure , Presenilins/antagonists & inhibitors , Presenilins/chemistry , Protein Conformation , Renin/antagonists & inhibitors , Renin/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL