Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Med Chem ; 67(2): 1447-1459, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38198520

ABSTRACT

Uveal melanoma (UM) is the most common primary intraocular malignancy in the adult eye. Despite the aggressive local management of primary UM, the development of metastases is common with no effective treatment options for metastatic disease. Genetic analysis of UM samples reveals the presence of mutually exclusive activating mutations in the Gq alpha subunits GNAQ and GNA11. One of the key downstream targets of the constitutively active Gq alpha subunits is the protein kinase C (PKC) signaling pathway. Herein, we describe the discovery of darovasertib (NVP-LXS196), a potent pan-PKC inhibitor with high whole kinome selectivity. The lead series was optimized for kinase and off target selectivity to afford a compound that is rapidly absorbed and well tolerated in preclinical species. LXS196 is being investigated in the clinic as a monotherapy and in combination with other agents for the treatment of uveal melanoma (UM), including primary UM and metastatic uveal melanoma (MUM).


Subject(s)
Melanoma , Uveal Neoplasms , Adult , Humans , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Melanoma/drug therapy , Melanoma/pathology , Uveal Neoplasms/drug therapy , Uveal Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mutation
2.
Blood Cancer J ; 12(8): 118, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35973981

ABSTRACT

We here defined the impacts of γ-secretase inhibitors (GSIs) on T-cell-dependent BCMA-specific multiple myeloma (MM) cell lysis and immunomodulatory effects induced by bispecific antibodies (BisAbs). GSIs-induced membrane BCMA (mBCMA) accumulation reached near maximum within 4 h and sustained over 42h-study period on MM cell lines and patient MM cells. GSIs, i.e., 2 nM LY-411575 or 1 µM DAPT, robustly increased mBCMA densities on CD138+ but not CD3+ patient cells, concomitantly with minimum soluble/shed BCMA (sBCMA) in 1 day-culture supernatants. In ex vivo MM-T-cell co-cultures, GSIs overcame sBCMA-inhibited MM cell lysis and further enhanced autologous patient MM cell lysis induced by BCMAxCD3 BisAbs, accompanied by significantly enhanced cytolytic markers (CD107a, IFNγ, IL2, and TNFα) in patient T cells. In longer 7 day-co-cultures, LY-411575 minimally affected BCMAxCD3 BisAb (PL33)-induced transient expression of checkpoint (PD1, TIGIT, TIM3, LAG3) and co-stimulatory (41BB, CD28) proteins, as well as time-dependent increases in % effector memory/central memory subsets and CD8/CD4 ratios in patient T cells. Importantly, LY41157 rapidly cleared sBCMA from circulation of MM-bearing NSG mice reconstituted with human T cells and significantly enhanced anti-MM efficacy of PL33 with prolonged host survival. Taken together, these results further support ongoing combination BCMA-targeting immunotherapies with GSI clinical studies to improve patient outcome.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Amyloid Precursor Protein Secretases , Animals , Antibodies, Bispecific/therapeutic use , B-Cell Maturation Antigen , Humans , Mice , Multiple Myeloma/drug therapy , T-Lymphocytes
3.
Front Immunol ; 13: 909979, 2022.
Article in English | MEDLINE | ID: mdl-35990699

ABSTRACT

CD3-engaging bispecific antibodies (BsAbs) enable the formation of an immune synapse between T cells and tumor cells, resulting in robust target cell killing not dependent on a preexisting tumor specific T cell receptor. While recent studies have shed light on tumor cell-specific factors that modulate BsAb sensitivity, the T cell-intrinsic determinants of BsAb efficacy and response durability are poorly understood. To better clarify the genes that shape BsAb-induced T cell responses, we conducted targeted analyses and a large-scale unbiased in vitro CRISPR/Cas9-based screen to identify negative regulators of BsAb-induced T cell proliferation. These analyses revealed that CD8+ T cells are dependent on CD4+ T cell-derived signaling factors in order to achieve sustained killing in vitro. Moreover, the mammalian target of rapamycin (mTOR) pathway and several other candidate genes were identified as intrinsic regulators of BsAb-induced T cell proliferation and/or activation, highlighting promising approaches to enhancing the utility of these potent therapeutics.


Subject(s)
Antibodies, Bispecific , Neoplasms , Antibodies, Bispecific/pharmacology , Antibody Formation , Humans , Lymphocyte Activation/genetics , Receptors, Antigen, T-Cell
4.
Bioorg Med Chem Lett ; 18(17): 4880-4, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18678489

ABSTRACT

This communication describes the discovery of a novel series of Aurora kinase inhibitors. Key SAR and critical binding elements are discussed. Some of the more advanced analogues potently inhibit cellular proliferation and induce phenotypes consistent with Aurora kinase inhibition. In particular, compound 21 (SNS-314) is a potent and selective Aurora kinase inhibitor that exhibits significant activity in pre-clinical in vivo tumor models.


Subject(s)
Neoplasms, Experimental/drug therapy , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aurora Kinases , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Mice , Neoplasm Transplantation , Neoplasms, Experimental/enzymology , Quinazolines/chemistry , Structure-Activity Relationship
5.
Nat Biotechnol ; 21(3): 308-14, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12563278

ABSTRACT

Cysteine aspartyl protease-3 (caspase-3) is a mediator of apoptosis and a therapeutic target for a wide range of diseases. Using a dynamic combinatorial technology, 'extended tethering', we identified unique nonpeptidic inhibitors for this enzyme. Extended tethering allowed the identification of ligands that bind to discrete regions of caspase-3 and also helped direct the assembly of these ligands into small-molecule inhibitors. We first designed a small-molecule 'extender' that irreversibly alkylates the cysteine residue of caspase-3 and also contains a thiol group. The modified protein was then screened against a library of disulfide-containing small-molecule fragments. Mass-spectrometry was used to identify ligands that bind noncovalently to the protein and that also form a disulfide linkage with the extender. Linking the selected fragments with binding elements from the extenders generates reversible, tight-binding molecules that are druglike and distinct from known inhibitors. One molecule derived from this approach inhibited apoptosis in cells.


Subject(s)
Caspase Inhibitors , Caspases/chemistry , Combinatorial Chemistry Techniques/methods , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Jurkat Cells/drug effects , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/classification , Humans , Jurkat Cells/metabolism , Mass Spectrometry/methods , Models, Molecular , Peptide Library
6.
Article in English | MEDLINE | ID: mdl-16511067

ABSTRACT

Caspase-1 is a key endopeptidase responsible for the post-translational processing of the IL-1beta and IL-18 cytokines and small-molecule inhibitors that modulate the activity of this enzyme are predicted to be important therapeutic treatments for many inflammatory diseases. A fragment-assembly approach, accompanied by structural analysis, was employed to generate caspase-1 inhibitors. With the aid of Tethering with extenders (small molecules that bind to the active-site cysteine and contain a free thiol), two novel fragments that bound to the active site and made a disulfide bond with the extender were identified by mass spectrometry. Direct linking of each fragment to the extender generated submicromolar reversible inhibitors that significantly reduced secretion of IL-1beta but not IL-6 from human peripheral blood mononuclear cells. Thus, Tethering with extenders facilitated rapid identification and synthesis of caspase-1 inhibitors with cell-based activity and subsequent structural analyses provided insights into the enzyme's ability to accommodate different inhibitor-binding modes in the active site.


Subject(s)
Caspase Inhibitors , Combinatorial Chemistry Techniques/methods , Cysteine Proteinase Inhibitors/chemistry , Binding Sites/drug effects , Caspase 1/chemistry , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Drug Design , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/blood , Interleukin-6/antagonists & inhibitors , Interleukin-6/blood , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/metabolism , Models, Molecular , Protein Binding/drug effects , Solubility
7.
J Med Chem ; 45(23): 5005-22, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12408711

ABSTRACT

The design, synthesis, and in vitro activities of a series of potent and selective small-molecule inhibitors of caspase-3 are described. From extended tethering, a salicylic acid fragment was identified as having binding affinity for the S(4) pocket of caspase-3. X-ray crystallography and molecular modeling of the initial tethering hit resulted in the synthesis of 4, which reversibly inhibited caspase-3 with a K(i) = 40 nM. Further optimization led to the identification of a series of potent and selective inhibitors with K(i) values in the 20-50 nM range. One of the most potent compounds in this series, 66b, inhibited caspase-3 with a K(i) = 20 nM and selectivity of 8-500-fold for caspase-3 vs a panel of seven caspases (1, 2, and 4-8). A high-resolution X-ray cocrystal structure of 4 and 66b supports the predicted binding modes of our compounds with caspase-3.


Subject(s)
Aspartic Acid/chemical synthesis , Caspase Inhibitors , Enzyme Inhibitors/chemical synthesis , Salicylates/chemical synthesis , Sulfonamides/chemical synthesis , Aspartic Acid/analogs & derivatives , Aspartic Acid/chemistry , Benzoxazoles/chemical synthesis , Benzoxazoles/chemistry , Binding Sites , Caspase 3 , Caspases/chemistry , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Fluorenes/chemical synthesis , Fluorenes/chemistry , Humans , Protein Binding , Salicylates/chemistry , Sulfonamides/chemistry , Thiophenes/chemical synthesis , Thiophenes/chemistry
8.
Cancer Chemother Pharmacol ; 65(4): 707-17, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19649632

ABSTRACT

PURPOSE: The Aurora family of serine/threonine kinases (Aurora-A, Aurora-B, and Aurora-C) plays a key role in cells orderly progression through mitosis. Elevated expression levels of Aurora kinases have been detected in a high percentage of melanoma, colon, breast, ovarian, gastric, and pancreatic tumors. We characterized the biological and pharmacological properties of SNS-314, an ATP-competitive, selective, and potent inhibitor of Aurora kinases. METHODS: We studied the biochemical potency and selectivity of SNS-314 to inhibit Aurora kinases A, B, and C. The inhibition of cellular proliferation induced by SNS-314 was evaluated in a broad range of tumor cell lines and correlated to inhibition of histone H3 phosphorylation, inhibition of cell-cycle progression, increase in nuclear content and cell size, loss of viability, and induction of apoptosis. The dose and administration schedule of SNS-314 was optimized for in vivo efficacy in mouse xenograft models of human cancer. RESULTS: In the HCT116 human colon cancer xenograft model, administration of 50 and 100 mg/kg SNS-314 led to dose-dependent inhibition of histone H3 phosphorylation for at least 10 h, indicating effective Aurora-B inhibition in vivo. HCT116 tumors from animals treated with SNS-314 showed potent and sustained responses including reduction of phosphorylated histone H3 levels, increased caspase-3 and appearance of increased nuclear size. The compound showed significant tumor growth inhibition in a dose-dependent manner under a variety of dosing schedules including weekly, bi-weekly, and 5 days on/9 days off. CONCLUSIONS: SNS-314 is a potent small-molecule inhibitor of Aurora kinases developed as a novel anti-cancer therapeutic agent for the treatment of diverse human malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/prevention & control , Phenylurea Compounds/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Thiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Aurora Kinase A , Aurora Kinase B , Aurora Kinase C , Aurora Kinases , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , HCT116 Cells , HT29 Cells , HeLa Cells , Histones/metabolism , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Phenylurea Compounds/chemistry , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Thiazoles/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Bioorg Med Chem Lett ; 16(3): 559-62, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16274992

ABSTRACT

Disulfide Tethering was applied to the active site of human caspase-1, resulting in the discovery of a novel, tricyclic molecular fragment that selectively binds in S4. This fragment was developed into a class of potent inhibitors of human caspase-1. Several key analogues determined the optimal distance of the tricycle from the catalytic residues, the relative importance of various features of the tricycle, and the importance of the linker.


Subject(s)
Caspase Inhibitors , Enzyme Inhibitors/chemical synthesis , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Binding Sites , Caspase 1/chemistry , Catalysis , Enzyme Inhibitors/pharmacology , Humans , Structure-Activity Relationship
10.
Proc Natl Acad Sci U S A ; 101(34): 12461-6, 2004 Aug 24.
Article in English | MEDLINE | ID: mdl-15314233

ABSTRACT

Allosteric regulation of proteins by conformational change is a primary means of biological control. Traditionally it has been difficult to identify and characterize novel allosteric sites and ligands that freeze these conformational states. We present a site-directed approach using Tethering for trapping inhibitory small molecules at sites away from the active site by reversible disulfide bond formation. We screened a library of 10,000 thiol-containing compounds against accessible cysteines of two members of the caspase family of proteases, caspase-3 and -7. We discovered a previously unreported and conserved allosteric site in a deep cavity at the dimer interface 14 A from the active site. This site contains a natural cysteine that, when disulfide-bonded with either of two specific compounds, inactivates these proteases. The allosteric site is functionally coupled to the active site, such that binding of the compounds at the allosteric site prevents peptide binding at the active site. The x-ray crystal structures of caspase-7 bound by either compound demonstrates that they inhibit caspase-7 by trapping a zymogen-like conformation. This approach may be useful to identify new allosteric sites from natural or engineered cysteines, to study allosteric transitions in proteins, and to nucleate drug discovery efforts.


Subject(s)
Allosteric Site , Caspases/chemistry , Protein Structure, Quaternary , Allosteric Regulation , Amides/chemistry , Binding Sites , Caspase Inhibitors , Caspases/metabolism , Crystallography, X-Ray , Dimerization , Disulfides/chemistry , Models, Molecular , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL