Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Transfusion ; 62(11): 2334-2348, 2022 11.
Article in English | MEDLINE | ID: mdl-36239134

ABSTRACT

BACKGROUND: There are two FDA-approved anti-CD38 monoclonal antibodies for treatment of multiple myeloma: isatuximab and daratumumab. Owing to expression of CD38 on reagent red blood cells (RBCs), these antibodies interfere with indirect antiglobulin tests (IATs). We sought to understand differences in such interference by performing binding experiments. STUDY DESIGN AND METHODS: In vitro experiments to compare the binding to RBCs of isatuximab and daratumumab alone or in the presence of a mouse anti-human CD38 antibody (HB-7 or AT13/5) or a nicotinamide adenine dinucleotide-analog CD38 inhibitor were performed and quantified by flow cytometry, imaging, mass spectrometry, surface plasmon resonance, and LigandTracer technologies. Serologic testing was performed on plasma samples spiked with isatuximab or daratumumab. RESULTS: CD38 expressed on RBCs can be directly bound by daratumumab, whereas isatuximab requires a co-factor, such as HB-7, AT13/5, or a CD38 inhibitor, suggesting that the isatuximab epitope on RBCs is masked in vitro. Daratumumab samples more frequently showed interference and had stronger reactions than isatuximab samples. Dithiothreitol treatment was equally effective in mitigating the interference caused by either drug. DISCUSSION: Both isatuximab and daratumumab interfere with IATs but at different magnitudes, reflecting distinct binding to CD38 on RBCs. From the binding studies, we conclude that the isatuximab epitope on RBCs is masked in vitro and binding requires a certain CD38 conformation or co-factor. This circumstance may explain why interference is seen only in a subset of patients receiving isatuximab when compared with interference seen in most patients on daratumumab therapy.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Neuroblastoma , Mice , Animals , ADP-ribosyl Cyclase 1 , Epitope Mapping , Antibodies, Monoclonal , Multiple Myeloma/therapy , Antineoplastic Agents/therapeutic use , Neuroblastoma/drug therapy , Epitopes
2.
Vaccine ; 37(42): 6208-6220, 2019 09 30.
Article in English | MEDLINE | ID: mdl-31493950

ABSTRACT

Seasonal influenza vaccines represent a positive intervention to limit the spread of the virus and protect public health. Yet continual influenza evolution and its ability to evade immunity pose a constant threat. For these reasons, vaccines with improved potency and breadth of protection remain an important need. We previously developed a next-generation influenza vaccine that displays the trimeric influenza hemagglutinin (HA) on a ferritin nanoparticle (NP) to optimize its presentation. Similar to other vaccines, HA-nanoparticle vaccine efficacy is increased by the inclusion of adjuvants during immunization. To identify the optimal adjuvants to enhance influenza immunity, we systematically analyzed TLR agonists for their ability to elicit immune responses. HA-NPs were compatible with nearly all adjuvants tested, including TLR2, TLR4, TLR7/8, and TLR9 agonists, squalene oil-in-water mixtures, and STING agonists. In addition, we chemically conjugated TLR7/8 and TLR9 ligands directly to the HA-ferritin nanoparticle. These TLR agonist-conjugated nanoparticles induced stronger antibody responses than nanoparticles alone, which allowed the use of a 5000-fold-lower dose of adjuvant than traditional admixtures. One candidate, the oil-in-water adjuvant AF03, was also tested in non-human primates and showed strong induction of neutralizing responses against both matched and heterologous H1N1 viruses. These data suggest that AF03, along with certain TLR agonists, enhance strong neutralizing antibody responses following influenza vaccination and may improve the breadth, potency, and ultimately vaccine protection in humans.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Neutralizing/immunology , Influenza Vaccines/immunology , Adjuvants, Immunologic/chemistry , Animals , Female , HEK293 Cells , Hemagglutination Inhibition Tests , Hemagglutinins , Humans , Macaca mulatta , Mice, Inbred BALB C , Nanoparticles , Toll-Like Receptors/agonists
3.
SLAS Discov ; 23(3): 264-273, 2018 03.
Article in English | MEDLINE | ID: mdl-29336194

ABSTRACT

CD73/Ecto-5'-nucleotidase is a membrane-tethered ecto-enzyme that works in tandem with CD39 to convert extracellular adenosine triphosphate (ATP) into adenosine. CD73 is highly expressed on various types of cancer cells and on infiltrating suppressive immune cells, leading to an elevated concentration of adenosine in the tumor microenvironment, which elicits a strong immunosuppressive effect. In preclinical studies, targeting CD73 with anti-CD73 antibody results in favorable antitumor effects. Despite initial studies using antibodies, inhibition of CD73 catalytic activity using small-molecule inhibitors may be more effective in lowering extracellular adenosine due to better tumor penetration and distribution. To screen small-molecule libraries, we explored multiple approaches, including colorimetric and fluorescent biochemical assays, and due to some limitations with these assays, we developed a mass spectrometry (MS)-based assay. Only the MS-based assay offers the sensitivity and dynamic range required for screening small-molecule libraries at a substrate concentration close to the Km value of substrate and for evaluating the mode of binding of screening hits. To achieve a throughput suitable for high-throughput screening (HTS), we developed a RapidFire-tandem mass spectrometry (RF-MS/MS)-based multiplex assay. This assay allowed a large diverse compound library to be screened at a speed of 1536 reactions per 40-50 min.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Assay/methods , Cell Line , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , HEK293 Cells , Humans , Mice , Tandem Mass Spectrometry/methods
4.
Cancer Cell ; 28(6): 773-784, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26678339

ABSTRACT

Heterozygous mutation of IDH1 in cancers modifies IDH1 enzymatic activity, reprogramming metabolite flux and markedly elevating 2-hydroxyglutarate (2-HG). Here, we found that 2-HG depletion did not inhibit growth of several IDH1 mutant solid cancer types. To identify other metabolic therapeutic targets, we systematically profiled metabolites in endogenous IDH1 mutant cancer cells after mutant IDH1 inhibition and discovered a profound vulnerability to depletion of the coenzyme NAD+. Mutant IDH1 lowered NAD+ levels by downregulating the NAD+ salvage pathway enzyme nicotinate phosphoribosyltransferase (Naprt1), sensitizing to NAD+ depletion via concomitant nicotinamide phosphoribosyltransferase (NAMPT) inhibition. NAD+ depletion activated the intracellular energy sensor AMPK, triggered autophagy, and resulted in cytotoxicity. Thus, we identify NAD+ depletion as a metabolic susceptibility of IDH1 mutant cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Isocitrate Dehydrogenase/genetics , Mutation , NAD/deficiency , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/drug effects , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cytokines/metabolism , Energy Metabolism/drug effects , Enzyme Activation , Female , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Glutarates/metabolism , HEK293 Cells , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/metabolism , Metabolomics/methods , Mice, SCID , Molecular Targeted Therapy , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/metabolism , Signal Transduction/drug effects , Spheroids, Cellular , Time Factors , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Biol Chem ; 290(2): 762-74, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25391653

ABSTRACT

Cancer-associated point mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) confer a neomorphic enzymatic activity: the reduction of α-ketoglutarate to d-2-hydroxyglutaric acid, which is proposed to act as an oncogenic metabolite by inducing hypermethylation of histones and DNA. Although selective inhibitors of mutant IDH1 and IDH2 have been identified and are currently under investigation as potential cancer therapeutics, the mechanistic basis for their selectivity is not yet well understood. A high throughput screen for selective inhibitors of IDH1 bearing the oncogenic mutation R132H identified compound 1, a bis-imidazole phenol that inhibits d-2-hydroxyglutaric acid production in cells. We investigated the mode of inhibition of compound 1 and a previously published IDH1 mutant inhibitor with a different chemical scaffold. Steady-state kinetics and biophysical studies show that both of these compounds selectively inhibit mutant IDH1 by binding to an allosteric site and that inhibition is competitive with respect to Mg(2+). A crystal structure of compound 1 complexed with R132H IDH1 indicates that the inhibitor binds at the dimer interface and makes direct contact with a residue involved in binding of the catalytically essential divalent cation. These results show that targeting a divalent cation binding residue can enable selective inhibition of mutant IDH1 and suggest that differences in magnesium binding between wild-type and mutant enzymes may contribute to the inhibitors' selectivity for the mutant enzyme.


Subject(s)
Drug Discovery , Enzyme Inhibitors/chemistry , Isocitrate Dehydrogenase/chemistry , Neoplasms/drug therapy , Allosteric Site , Crystallography, X-Ray , DNA Methylation/genetics , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Escherichia coli , Gene Expression Regulation, Neoplastic , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/biosynthesis , Isocitrate Dehydrogenase/genetics , Magnesium/chemistry , Mutant Proteins/chemistry , Mutant Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Conformation
6.
Chem Commun (Camb) ; 47(4): 1246-8, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21103558

ABSTRACT

Here we report the synthesis of monofunctional PEGylated amide ligands that were used to prepare bioactivable quantum dots of a 20 nm diameter with a controlled mean number of the covalently grafted ligands. They are stable in aqueous medium of high salinity including a large pH domain.


Subject(s)
Polyethylene Glycols/chemistry , Quantum Dots , Micelles , Peptides/chemistry , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL