Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cancer Res ; 83(12): 1941-1952, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37140427

ABSTRACT

Major advances have been made in the field of precision medicine for treating cancer. However, many open questions remain that need to be answered to realize the goal of matching every patient with cancer to the most efficacious therapy. To facilitate these efforts, we have developed CellMinerCDB: National Center for Advancing Translational Sciences (NCATS; https://discover.nci.nih.gov/rsconnect/cellminercdb_ncats/), which makes available activity information for 2,675 drugs and compounds, including multiple nononcology drugs and 1,866 drugs and compounds unique to the NCATS. CellMinerCDB: NCATS comprises 183 cancer cell lines, with 72 unique to NCATS, including some from previously understudied tissues of origin. Multiple forms of data from different institutes are integrated, including single and combination drug activity, DNA copy number, methylation and mutation, transcriptome, protein levels, histone acetylation and methylation, metabolites, CRISPR, and miscellaneous signatures. Curation of cell lines and drug names enables cross-database (CDB) analyses. Comparison of the datasets is made possible by the overlap between cell lines and drugs across databases. Multiple univariate and multivariate analysis tools are built-in, including linear regression and LASSO. Examples have been presented here for the clinical topoisomerase I (TOP1) inhibitors topotecan and irinotecan/SN-38. This web application provides both substantial new data and significant pharmacogenomic integration, allowing exploration of interrelationships. SIGNIFICANCE: CellMinerCDB: NCATS provides activity information for 2,675 drugs in 183 cancer cell lines and analysis tools to facilitate pharmacogenomic research and to identify determinants of response.


Subject(s)
National Center for Advancing Translational Sciences (U.S.) , Neoplasms, Basal Cell , United States , Humans , Pharmacogenetics , Cell Line, Tumor , Databases, Factual , Irinotecan , Internet
2.
ACS Pharmacol Transl Sci ; 5(6): 445-447, 2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35702394

ABSTRACT

The SARS-CoV-2 main protease is among the most attractive targets for the development of therapeutic interventions for COVID-19. Successful candidate agents will not only possess potent on-target activity versus SARS-CoV-2 Mpro but also minimal polypharmacology versus human cysteine proteases. This Viewpoint explores the activity profile of the first approved SARS-CoV-2 Mpro inhibitor (Nirmatrelvir) versus a panel of cysteine proteases and considers the therapeutic implications of the data.

3.
Mol Cancer Res ; 18(10): 1512-1521, 2020 10.
Article in English | MEDLINE | ID: mdl-32611550

ABSTRACT

O-GlcNAc transferase (OGT) is a nutrient-sensitive glycosyltransferase that is overexpressed in prostate cancer, the most common cancer in males. We recently developed a specific and potent inhibitor targeting this enzyme, and here, we report a synthetic lethality screen using this compound. Our screen identified pan-cyclin-dependent kinase (CDK) inhibitor AT7519 as lethal in combination with OGT inhibition. Follow-up chemical and genetic approaches identified CDK9 as the major target for synthetic lethality with OGT inhibition in prostate cancer cells. OGT expression is regulated through retention of the fourth intron in the gene and CDK9 inhibition blunted this regulatory mechanism. CDK9 phosphorylates carboxy-terminal domain (CTD) of RNA Polymerase II to promote transcription elongation. We show that OGT inhibition augments effects of CDK9 inhibitors on CTD phosphorylation and general transcription. Finally, the combined inhibition of both OGT and CDK9 blocked growth of organoids derived from patients with metastatic prostate cancer, but had minimal effects on normal prostate spheroids. We report a novel synthetic lethal interaction between inhibitors of OGT and CDK9 that specifically kills prostate cancer cells, but not normal cells. Our study highlights the potential of combining OGT inhibitors with other treatments to exploit cancer-specific vulnerabilities. IMPLICATIONS: The primary contribution of OGT to cell proliferation is unknown, and in this study, we used a compound screen to indicate that OGT and CDK9 collaborate to sustain a cancer cell-specific pro-proliferative program. A better understanding of how OGT and CDK9 cross-talk will refine our understanding of this novel synthetic lethal interaction.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cyclin-Dependent Kinase 9/metabolism , Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Synergism , Enzyme Inhibitors/administration & dosage , Humans , Male , Molecular Targeted Therapy , N-Acetylglucosaminyltransferases/metabolism , Piperidines/pharmacology , Prostatic Neoplasms/genetics , Protein Kinase Inhibitors/administration & dosage , Pyrazoles/pharmacology
4.
Mol Cancer Ther ; 19(7): 1520-1529, 2020 07.
Article in English | MEDLINE | ID: mdl-32371575

ABSTRACT

Despite a growing body of knowledge about the genomic landscape of Ewing sarcoma, translation of basic discoveries into targeted therapies and significant clinical gains has remained elusive. Recent insights have revealed that the oncogenic transcription factor EWS-FLI1 can impact Ewing sarcoma cellular metabolism, regulating expression of 3-phosphoglycerate dehydrogenase (PHGDH), the first enzyme in de novo serine synthesis. Here, we have examined the importance of serine metabolism in Ewing sarcoma tumorigenesis and evaluated the therapeutic potential of targeting serine metabolism in preclinical models of Ewing sarcoma. We show that PHGDH knockdown resulted in decreased Ewing sarcoma cell proliferation, especially under serine limitation, and significantly inhibited xenograft tumorigenesis in preclinical orthotopic models of Ewing sarcoma. In addition, the PHGDH inhibitor NCT-503 caused a dose-dependent decrease in cellular proliferation. Moreover, we report a novel drug combination in which nicotinamide phosphoribosyltransferase (NAMPT) inhibition, which blocks production of the PHGDH substrate NAD+, synergized with NCT-503 to abolish Ewing sarcoma cell proliferation and tumor growth. Furthermore, we show that serine deprivation inhibited Ewing sarcoma cell proliferation and tumorigenesis, indicating that Ewing sarcoma cells depend on exogenous serine in addition to de novo serine synthesis. Our findings suggest that serine metabolism is critical for Ewing sarcoma tumorigenesis, and that targeting metabolic dependencies should be further investigated as a potential therapeutic strategy for Ewing sarcoma. In addition, the combination strategy presented herein may have broader clinical applications in other PHGDH-overexpressing cancers as well.


Subject(s)
Bone Neoplasms/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Phosphoglycerate Dehydrogenase/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/pathology , Serine/metabolism , Animals , Apoptosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Female , Humans , Mice , Mice, SCID , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Nucleic Acids Res ; 48(10): 5656-5669, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32329777

ABSTRACT

Intron detention in precursor RNAs serves to regulate expression of a substantial fraction of genes in eukaryotic genomes. How detained intron (DI) splicing is controlled is poorly understood. Here, we show that a ubiquitous post-translational modification called O-GlcNAc, which is thought to integrate signaling pathways as nutrient conditions fluctuate, controls detained intron splicing. Using specific inhibitors of the enzyme that installs O-GlcNAc (O-GlcNAc transferase, or OGT) and the enzyme that removes O-GlcNAc (O-GlcNAcase, or OGA), we first show that O-GlcNAc regulates splicing of the highly conserved detained introns in OGT and OGA to control mRNA abundance in order to buffer O-GlcNAc changes. We show that OGT and OGA represent two distinct paradigms for how DI splicing can control gene expression. We also show that when DI splicing of the O-GlcNAc-cycling genes fails to restore O-GlcNAc homeostasis, there is a global change in detained intron levels. Strikingly, almost all detained introns are spliced more efficiently when O-GlcNAc levels are low, yet other alternative splicing pathways change minimally. Our results demonstrate that O-GlcNAc controls detained intron splicing to tune system-wide gene expression, providing a means to couple nutrient conditions to the cell's transcriptional regime.


Subject(s)
Acetylglucosamine/metabolism , Glycoside Hydrolases/genetics , Introns , N-Acetylglucosaminyltransferases/genetics , RNA Splicing , Cell Line , Glycoside Hydrolases/metabolism , HEK293 Cells , Humans , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , RNA Splicing Factors/metabolism , RNA, Messenger/metabolism , RNA-Seq
6.
PLoS One ; 15(4): e0226661, 2020.
Article in English | MEDLINE | ID: mdl-32240171

ABSTRACT

CD47 is an immune checkpoint protein that downregulates both the innate and adaptive anti-tumor immune response via its counter receptor SIRPα. Biologics, including humanized CD47 monoclonal antibodies and decoy SIRPα receptors, that block the SIRPα-CD47 interaction, are currently being developed as cancer immunotherapy agents. However, adverse side effects and limited penetration of tumor tissue associated with their structure and large size may impede their clinical application. We recently developed a quantitative high throughput screening assay platform to identify small molecules that disrupt the binding of SIRPα and CD47 as an alternative approach to these protein-based therapeutics. Here, we report on the development and optimization of a cell-based binding assay to validate active small molecules from our biochemical screening effort. This assay has a low volume, high capacity homogenous format that relies on laser scanning cytometry (LSC) and associated techniques to enhance signal to noise measurement of cell surface binding. The LSC assay is specific, concentration dependent, and validated for the two major human SIRPα variants (V1 and V2), with results that parallel those of our biochemical data as well as published studies. We also utilized the LSC assay to confirm published studies showing that the inhibition of amino-terminal pyroglutamate formation on CD47 using the glutaminyl cyclase inhibitor SEN177 disrupts SIRPα binding. The SIRPα-CD47 interaction could be quantitatively measured in live and fixed tumor cells. Use of fixed cells reduces the burden of cell maintenance and provides stable cell standards to control for inter- and intra-assay variations. We also demonstrate the utility of the assay to characterize the activity of the first reported small molecule antagonists of the SIRPα-CD47 interaction. This assay will support the screening of thousands of compounds to identify or validate active small molecules as hits, develop structure activity relationships and assist in the optimization of hits to leads by a typical iterative medicinal chemistry campaign.


Subject(s)
Adaptive Immunity/drug effects , Antigens, Differentiation/genetics , CD47 Antigen/genetics , Neoplasms/drug therapy , Receptors, Immunologic/genetics , Small Molecule Libraries/pharmacology , Adaptive Immunity/genetics , Aminoacyltransferases/antagonists & inhibitors , Aminoacyltransferases/chemistry , Antigens, Differentiation/chemistry , CD47 Antigen/chemistry , Drug Development , Gene Expression Regulation, Neoplastic/drug effects , High-Throughput Screening Assays/methods , Humans , Immunotherapy/methods , Jurkat Cells , Laser Scanning Cytometry , Ligands , Medical Oncology/trends , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Phagocytosis/drug effects , Protein Interaction Maps/genetics , Receptors, Immunologic/chemistry , Small Molecule Libraries/chemistry
7.
Sci Transl Med ; 11(519)2019 11 20.
Article in English | MEDLINE | ID: mdl-31748226

ABSTRACT

Diffuse midline gliomas (DMGs) are universally lethal malignancies occurring chiefly during childhood and involving midline structures of the central nervous system, including thalamus, pons, and spinal cord. These molecularly related cancers are characterized by high prevalence of the histone H3K27M mutation. In search of effective therapeutic options, we examined multiple DMG cultures in sequential quantitative high-throughput screens (HTS) of 2706 approved and investigational drugs. This effort generated 19,936 single-agent dose responses that inspired a series of HTS-enabled drug combination assessments encompassing 9195 drug-drug examinations. Top combinations were validated across patient-derived cell cultures representing the major DMG genotypes. In vivo testing in patient-derived xenograft models validated the combination of the multi-histone deacetylase (HDAC) inhibitor panobinostat and the proteasome inhibitor marizomib as a promising therapeutic approach. Transcriptional and metabolomic surveys revealed substantial alterations to key metabolic processes and the cellular unfolded protein response after treatment with panobinostat and marizomib. Mitigation of drug-induced cytotoxicity and basal mitochondrial respiration with exogenous application of nicotinamide mononucleotide (NMN) or exacerbation of these phenotypes when blocking nicotinamide adenine dinucleotide (NAD+) production via nicotinamide phosphoribosyltransferase (NAMPT) inhibition demonstrated that metabolic catastrophe drives the combination-induced cytotoxicity. This study provides a comprehensive single-agent and combinatorial drug screen for DMG and identifies concomitant HDAC and proteasome inhibition as a promising therapeutic strategy that underscores underrecognized metabolic vulnerabilities in DMG.


Subject(s)
Brain Neoplasms/drug therapy , Drug Evaluation, Preclinical , Glioma/drug therapy , High-Throughput Screening Assays/methods , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Stem Neoplasms/drug therapy , Cell Death , Cell Line, Tumor , Drug Synergism , Female , Glioma/genetics , Glioma/metabolism , Humans , Lactones/pharmacology , Lactones/therapeutic use , Male , Metabolomics , Mice , Panobinostat/pharmacology , Panobinostat/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Reproducibility of Results , Sequence Analysis, RNA , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
8.
J Am Chem Soc ; 140(42): 13542-13545, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30285435

ABSTRACT

Reversible glycosylation of nuclear and cytoplasmic proteins is an important regulatory mechanism across metazoans. One enzyme, O-linked N-acetylglucosamine transferase (OGT), is responsible for all nucleocytoplasmic glycosylation and there is a well-known need for potent, cell-permeable inhibitors to interrogate OGT function. Here we report the structure-based evolution of OGT inhibitors culminating in compounds with low nanomolar inhibitory potency and on-target cellular activity. In addition to disclosing useful OGT inhibitors, the structures we report provide insight into how to inhibit glycosyltransferases, a family of enzymes that has been notoriously refractory to inhibitor development.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , HCT116 Cells , HEK293 Cells , Humans , Molecular Docking Simulation , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
9.
Clin Cancer Res ; 23(23): 7301-7311, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28899971

ABSTRACT

Purpose: Although many cancers are showing remarkable responses to targeted therapies, pediatric sarcomas, including Ewing sarcoma, remain recalcitrant. To broaden the therapeutic landscape, we explored the in vitro response of Ewing sarcoma cell lines against a large collection of investigational and approved drugs to identify candidate combinations.Experimental Design: Drugs displaying activity as single agents were evaluated in combinatorial (matrix) format to identify highly active, synergistic drug combinations, and combinations were subsequently validated in multiple cell lines using various agents from each class. Comprehensive metabolomic and proteomic profiling was performed to better understand the mechanism underlying the synergy. Xenograft experiments were performed to determine efficacy and in vivo mechanism.Results: Several promising candidates emerged, including the combination of small-molecule PARP and nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, a rational combination as NAMPTis block the rate-limiting enzyme in the production of nicotinamide adenine dinucleotide (NAD+), a necessary substrate of PARP. Mechanistic drivers of the synergistic cell killing phenotype of these combined drugs included depletion of NMN and NAD+, diminished PAR activity, increased DNA damage, and apoptosis. Combination PARPis and NAMPTis in vivo resulted in tumor regression, delayed disease progression, and increased survival.Conclusions: These studies highlight the potential of these drugs as a possible therapeutic option in treating patients with Ewing sarcoma. Clin Cancer Res; 23(23); 7301-11. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Sarcoma, Ewing/drug therapy , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytokines/metabolism , Drug Screening Assays, Antitumor/methods , Drug Synergism , Enzyme Inhibitors/administration & dosage , Female , Humans , Kaplan-Meier Estimate , Mice, SCID , Nicotinamide Phosphoribosyltransferase/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Tumor Burden/drug effects
10.
Nature ; 549(7673): 533-537, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959975

ABSTRACT

High-grade gliomas (HGG) are a devastating group of cancers, and represent the leading cause of brain tumour-related death in both children and adults. Therapies aimed at mechanisms intrinsic to glioma cells have translated to only limited success; effective therapeutic strategies will need also to target elements of the tumour microenvironment that promote glioma progression. Neuronal activity promotes the growth of a range of molecularly and clinically distinct HGG types, including adult and paediatric glioblastoma (GBM), anaplastic oligodendroglioma, and diffuse intrinsic pontine glioma (DIPG). An important mechanism that mediates this neural regulation of brain cancer is activity-dependent cleavage and secretion of the synaptic adhesion molecule neuroligin-3 (NLGN3), which promotes glioma proliferation through the PI3K-mTOR pathway. However, the necessity of NLGN3 for glioma growth, the proteolytic mechanism of NLGN3 secretion, and the further molecular consequences of NLGN3 secretion in glioma cells remain unknown. Here we show that HGG growth depends on microenvironmental NLGN3, identify signalling cascades downstream of NLGN3 binding in glioma, and determine a therapeutically targetable mechanism of secretion. Patient-derived orthotopic xenografts of paediatric GBM, DIPG and adult GBM fail to grow in Nlgn3 knockout mice. NLGN3 stimulates several oncogenic pathways, such as early focal adhesion kinase activation upstream of PI3K-mTOR, and induces transcriptional changes that include upregulation of several synapse-related genes in glioma cells. NLGN3 is cleaved from both neurons and oligodendrocyte precursor cells via the ADAM10 sheddase. ADAM10 inhibitors prevent the release of NLGN3 into the tumour microenvironment and robustly block HGG xenograft growth. This work defines a promising strategy for targeting NLGN3 secretion, which could prove transformative for HGG therapy.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Glioma/metabolism , Glioma/pathology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/metabolism , Adult , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cell Proliferation , Child , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glioma/genetics , Heterografts , Humans , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Neoplasm Transplantation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/pathology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Microenvironment
11.
Oncotarget ; 7(11): 12464-76, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26824323

ABSTRACT

Metabolic networks are highly connected and complex, but a single enzyme, O-GlcNAc transferase (OGT) can sense the availability of metabolites and also modify target proteins. We show that inhibition of OGT activity inhibits the proliferation of prostate cancer cells, leads to sustained loss of c-MYC and suppresses the expression of CDK1, elevated expression of which predicts prostate cancer recurrence (p=0.00179). Metabolic profiling revealed decreased glucose consumption and lactate production after OGT inhibition. This decreased glycolytic activity specifically sensitized prostate cancer cells, but not cells representing normal prostate epithelium, to inhibitors of oxidative phosphorylation (rotenone and metformin). Intra-cellular alanine was depleted upon OGT inhibitor treatment. OGT inhibitor increased the expression and activity of alanine aminotransferase (GPT2), an enzyme that can be targeted with a clinically approved drug, cycloserine. Simultaneous inhibition of OGT and GPT2 inhibited cell viability and growth rate, and additionally activated a cell death response. These combinatorial effects were predominantly seen in prostate cancer cells, but not in a cell-line derived from normal prostate epithelium. Combinatorial treatments were confirmed with two inhibitors against both OGT and GPT2. Taken together, here we report the reprogramming of energy metabolism upon inhibition of OGT activity, and identify synergistically lethal combinations that are prostate cancer cell specific.


Subject(s)
Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Glycosylation , Humans , Male , N-Acetylglucosaminyltransferases/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism
12.
Proc Natl Acad Sci U S A ; 112(40): 12480-5, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26396258

ABSTRACT

Adult T-cell leukemia (ATL) develops in individuals infected with human T-cell lymphotropic virus-1 (HTLV-1). Presently there is no curative therapy for ATL. HTLV-1-encoded protein Tax (transactivator from the X-gene region) up-regulates Bcl-xL (B-cell lymphoma-extra large) expression and activates interleukin-2 (IL-2), IL-9, and IL-15 autocrine/paracrine systems, resulting in amplified JAK/STAT signaling. Inhibition of JAK signaling reduces cytokine-dependent ex vivo proliferation of peripheral blood mononuclear cells (PBMCs) from ATL patients in smoldering/chronic stages. Currently, two JAK inhibitors are approved for human use. In this study, we examined activity of multiple JAK inhibitors in ATL cell lines. The selective JAK inhibitor ruxolitinib was examined in a high-throughput matrix screen combined with >450 potential therapeutic agents, and Bcl-2/Bcl-xL inhibitor navitoclax was identified as a strong candidate for multicomponent therapy. The combination was noted to strongly activate BAX (Bcl-2-associated X protein), effect mitochondrial depolarization, and increase caspase 3/7 activities that lead to cleavage of PARP (poly ADP ribose polymerase) and Mcl-1 (myeloid cell leukemia 1). Ruxolitinib and navitoclax independently demonstrated modest antitumor efficacy, whereas the combination dramatically lowered tumor burden and prolonged survival in an ATL murine model. This combination strongly blocked ex vivo proliferation of five ATL patients' PBMCs. These studies provide support for a therapeutic trial in patients with smoldering/chronic ATL using a drug combination that inhibits JAK signaling and antiapoptotic protein Bcl-xL.


Subject(s)
Interleukin-2/metabolism , Janus Kinases/metabolism , Leukemia-Lymphoma, Adult T-Cell/metabolism , STAT Transcription Factors/metabolism , bcl-X Protein/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Female , Flow Cytometry , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Janus Kinases/antagonists & inhibitors , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nitriles , Pyrazoles/pharmacology , Pyrimidines , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction/drug effects , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors
13.
ACS Chem Biol ; 10(6): 1392-7, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25751766

ABSTRACT

O-GlcNAc transferase (OGT) is an essential mammalian enzyme that regulates numerous cellular processes through the attachment of O-linked N-acetylglucosamine (O-GlcNAc) residues to nuclear and cytoplasmic proteins. Its targets include kinases, phosphatases, transcription factors, histones, and many other intracellular proteins. The biology of O-GlcNAc modification is still not well understood, and cell-permeable inhibitors of OGT are needed both as research tools and for validating OGT as a therapeutic target. Here, we report a small molecule OGT inhibitor, OSMI-1, developed from a high-throughput screening hit. It is cell-permeable and inhibits protein O-GlcNAcylation in several mammalian cell lines without qualitatively altering cell surface N- or O-linked glycans. The development of this molecule validates high-throughput screening approaches for the discovery of glycosyltransferase inhibitors, and further optimization of this scaffold may lead to yet more potent OGT inhibitors useful for studying OGT in animal models.


Subject(s)
Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , CHO Cells , Cell Membrane Permeability , Cricetulus , Enzyme Inhibitors/chemical synthesis , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Lectins/chemistry , Lectins/metabolism , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Small Molecule Libraries/chemical synthesis , Uridine Diphosphate/chemistry , Uridine Diphosphate/metabolism , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism
14.
Proc Natl Acad Sci U S A ; 111(6): 2349-54, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469833

ABSTRACT

The clinical development of drug combinations is typically achieved through trial-and-error or via insight gained through a detailed molecular understanding of dysregulated signaling pathways in a specific cancer type. Unbiased small-molecule combination (matrix) screening represents a high-throughput means to explore hundreds and even thousands of drug-drug pairs for potential investigation and translation. Here, we describe a high-throughput screening platform capable of testing compounds in pairwise matrix blocks for the rapid and systematic identification of synergistic, additive, and antagonistic drug combinations. We use this platform to define potential therapeutic combinations for the activated B-cell-like subtype (ABC) of diffuse large B-cell lymphoma (DLBCL). We identify drugs with synergy, additivity, and antagonism with the Bruton's tyrosine kinase inhibitor ibrutinib, which targets the chronic active B-cell receptor signaling that characterizes ABC DLBCL. Ibrutinib interacted favorably with a wide range of compounds, including inhibitors of the PI3K-AKT-mammalian target of rapamycin signaling cascade, other B-cell receptor pathway inhibitors, Bcl-2 family inhibitors, and several components of chemotherapy that is the standard of care for DLBCL.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , B-Lymphocytes/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Cell Line, Tumor , High-Throughput Screening Assays , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Phosphatidylinositol 3-Kinases/metabolism , Piperidines
15.
Bioorg Med Chem Lett ; 24(2): 630-5, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24360556

ABSTRACT

The structure-activity relationship (SAR) study of two chemotypes identified as inhibitors of the human NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (HPGD, 15-PGDH) was conducted. Top compounds from both series displayed potent inhibition (IC50 <50 nM), demonstrate excellent selectivity towards HPGD and potently induce PGE2 production in A549 lung cancer and LNCaP prostate cancer cells.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , NAD/antagonists & inhibitors , Cell Line, Tumor , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , NAD/metabolism , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 23(3): 682-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23273517

ABSTRACT

The importance of the trifluoromethyl group in the polypharmacological profile of nilotinib was investigated. Molecular editing of nilotinib led to the design, synthesis and biological evaluation of analogues where the trifluoromethyl group was replaced by a proton, fluorine and a methyl group. While these analogues were less active than nilotinib toward Abl, their activity toward Kit was comparable, with the monofluorinated analogue being the most active. Docking of nilotinib and of analogues 2a-c to the binding pocket of Abl and of Kit showed that the lack of shape complementarity in Kit is compensated by the stabilizing effect from its juxtamembrane region.


Subject(s)
Oncogene Proteins v-abl/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Catalytic Domain , Drug Design , Enzyme Activation/drug effects , Fluorine/chemistry , Methane/chemistry , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry
17.
Bioorg Med Chem ; 18(17): 6429-41, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20691600

ABSTRACT

Analogues of mitoQ and idebenone were synthesized to define the structural elements that support oxygen consumption in the mitochondrial respiratory chain. Eight analogues were prepared and fully characterized, then evaluated for their ability to support oxygen consumption in the mitochondrial respiratory chain. While oxygen consumption was strongly inhibited by mitoQ analogues 2-4 in a chain length-dependent manner, modification of idebenone by replacement of the quinone methoxy groups by methyl groups (analogues 6-8) reduced, but did not eliminate, oxygen consumption. Idebenone analogues 6-8 also displayed significant cytoprotective properties toward cultured mammalian cells in which glutathione had been depleted by treatment with diethyl maleate.


Subject(s)
Mitochondria/drug effects , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/pharmacology , Oxygen Consumption/drug effects , Ubiquinone/analogs & derivatives , Antioxidants/chemical synthesis , Antioxidants/pharmacology , Cytoprotection , Glutathione/metabolism , Humans , Mitochondria/metabolism , Ubiquinone/chemical synthesis , Ubiquinone/pharmacology
18.
J Org Chem ; 71(13): 5027-30, 2006 Jun 23.
Article in English | MEDLINE | ID: mdl-16776541

ABSTRACT

Tributyltin hydride-mediated cyclizations of 1-nitro-2-acetoxy-5-hexenes 7a-g having multiple substitutions on carbons 1 and 6 result in 2,3-substituted-1-acetoxycyclopentanes 1a-g. The substrates were prepared by nitroaldol reactions of silyloxyaldehydes followed by acetylation, desilylation, and oxidation to the acetoxynitroaldehydes 6a-e. Wittig olefination of aldehydes 6a-e then afforded substrates for the radical cyclizations. The overall scheme gave a diverse array of cyclopentanes, including gem-disubstituted cyclopentanes having substitution on three contiguous carbons.


Subject(s)
Cyclopentanes/chemical synthesis , Nitro Compounds/chemical synthesis , Alkenes/chemistry , Cyclization , Cyclopentanes/chemistry , Free Radicals/chemistry , Molecular Structure , Nitro Compounds/chemistry , Stereoisomerism , Trialkyltin Compounds/chemistry
19.
J Org Chem ; 70(24): 10117-20, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16292851

ABSTRACT

[reaction: see text] A synthesis of the glutarimide-derived metabolite of thalidomide, 5'-hydroxythalidomide (2), is described. The synthesis employed the lactone derivative of N-benzyloxycarbonyl (CBZ)-protected 4-hydroxyglutamic acid 12, which is prepared by a de novo route from diethyl acetamidomalonate. The reaction of 12 with 4-methoxybenzylamine gave the corresponding isoglutamine, which then provided the key CBZ-protected N-PMB-glutarimide 14 after dehydration. Deprotection of both the CBZ and PMB groups followed by phthalimidation and deacetylation of the 3-amino-5-acetoxyglutarimide 16 afforded 2.


Subject(s)
Phthalimides/chemical synthesis , Phthalimides/isolation & purification , Piperidones/chemical synthesis , Piperidones/isolation & purification , Thalidomide/chemistry , Humans , Molecular Structure , Phthalimides/chemistry , Piperidones/chemistry , Stereoisomerism , Thalidomide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...