Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Mol Ther ; 32(6): 1895-1916, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38549376

Malignant tumors are often associated with an immunosuppressive tumor microenvironment (TME), rendering most of them resistant to standard-of-care immune checkpoint inhibitors (CPIs). Signal transducer and activator of transcription 3 (STAT3), a ubiquitously expressed transcription factor, has well-defined immunosuppressive functions in several leukocyte populations within the TME. Since the STAT3 protein has been challenging to target using conventional pharmaceutical modalities, we investigated the feasibility of applying systemically delivered RNA interference (RNAi) agents to silence its mRNA directly in tumor-associated immune cells. In preclinical rodent tumor models, chemically stabilized acylated small interfering RNAs (siRNAs) selectively silenced Stat3 mRNA in multiple relevant cell types, reduced STAT3 protein levels, and increased cytotoxic T cell infiltration. In a murine model of CPI-resistant pancreatic cancer, RNAi-mediated Stat3 silencing resulted in tumor growth inhibition, which was further enhanced in combination with CPIs. To further exemplify the utility of RNAi for cancer immunotherapy, this technology was used to silence Cd274, the gene encoding the immune checkpoint protein programmed death-ligand 1 (PD-L1). Interestingly, silencing of Cd274 was effective in tumor models that are resistant to PD-L1 antibody therapy. These data represent the first demonstration of systemic delivery of RNAi agents to the TME and suggest applying this technology for immuno-oncology applications.


B7-H1 Antigen , RNA Interference , RNA, Small Interfering , STAT3 Transcription Factor , Tumor Microenvironment , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Animals , Mice , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Humans , Tumor Microenvironment/immunology , RNA, Small Interfering/genetics , Immunotherapy/methods , Drug Resistance, Neoplasm/genetics , Immune Checkpoint Inhibitors/pharmacology , Disease Models, Animal , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/genetics
2.
Cancer Prev Res (Phila) ; 8(9): 786-95, 2015 Sep.
Article En | MEDLINE | ID: mdl-26069204

Despite widespread use as well as epidemiologic indications, there have been no investigations into the effect of St. John's wort (SJW) extract on colorectal carcinogenesis in vivo. This study reports a systematic evaluation of the impact of dietary supplementation of SJW extract on azoxymethane-induced colorectal carcinogenesis in mice. Mice were fed with either AIN-93G (control) diet or SJW extract-supplemented diet (SJW diet) prior to azoxymethane treatment. SJW diet was found to significantly improve the overall survival of azoxymethane-treated mice. Pretreatment with the SJW diet significantly reduced body weight loss as well as decrease of serum albumin and cholesterol levels associated with azoxymethane-induced colorectal tumorigenesis. SJW diet-fed mice showed a significant decrease in tumor multiplicity along with a decrease in incidence of large tumors and a trend toward decreased total tumor volume in a dose-dependent manner. A short-term study, which examined the effect of SJW prior to rectal bleeding, also showed decrease in colorectal polyps in SJW diet-fed mice. Nuclear factor kappa B (NF-κB) and extracellular signal-regulated kinase (ERK1/2) pathways were attenuated by SJW administration. SJW extract resulted in early and continuous attenuation of these pathways in the colon epithelium of SJW diet-fed mice under both short-term and long-term treatment regimens. In conclusion, this study demonstrated the chemopreventive potential of SJW extract against colorectal cancer through attenuation of proinflammatory processes.


Anticarcinogenic Agents/therapeutic use , Carcinogenesis/drug effects , Colorectal Neoplasms/prevention & control , Hypericum/chemistry , Inflammation/drug therapy , Plant Extracts/therapeutic use , Animals , Azoxymethane/chemistry , Cell Transformation, Neoplastic/drug effects , Colon/pathology , Colorectal Neoplasms/drug therapy , Diet , Dietary Supplements , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides/chemistry , Signal Transduction/drug effects
3.
J Med Chem ; 56(5): 2110-24, 2013 Mar 14.
Article En | MEDLINE | ID: mdl-23445220

Protein arginine methyltransferases (PRMTs) play an important role in diverse biological processes. Among the nine known human PRMTs, PRMT3 has been implicated in ribosomal biosynthesis via asymmetric dimethylation of the 40S ribosomal protein S2 and in cancer via interaction with the DAL-1 tumor suppressor protein. However, few selective inhibitors of PRMTs have been discovered. We recently disclosed the first selective PRMT3 inhibitor, which occupies a novel allosteric binding site and is noncompetitive with both the peptide substrate and cofactor. Here we report comprehensive structure-activity relationship studies of this series, which resulted in the discovery of multiple PRMT3 inhibitors with submicromolar potencies. An X-ray crystal structure of compound 14u in complex with PRMT3 confirmed that this inhibitor occupied the same allosteric binding site as our initial lead compound. These studies provide the first experimental evidence that potent and selective inhibitors can be created by exploiting the allosteric binding site of PRMT3.


Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Allosteric Site/drug effects , Enzyme Inhibitors/chemical synthesis , Humans , Inhibitory Concentration 50 , Protein-Arginine N-Methyltransferases/chemistry , Ribosomal Proteins/metabolism , Structure-Activity Relationship , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , X-Ray Diffraction
4.
Structure ; 20(8): 1425-35, 2012 Aug 08.
Article En | MEDLINE | ID: mdl-22795084

PRMT3, a protein arginine methyltransferase, has been shown to influence ribosomal biosynthesis by catalyzing the dimethylation of the 40S ribosomal protein S2. Although PRMT3 has been reported to be a cytosolic protein, it has been shown to methylate histone H4 peptide (H4 1-24) in vitro. Here, we report the identification of a PRMT3 inhibitor (1-(benzo[d][1,2,3]thiadiazol-6-yl)-3-(2-cyclohexenylethyl)urea; compound 1) with IC50 value of 2.5 µM by screening a library of 16,000 compounds using H4 (1-24) peptide as a substrate. The crystal structure of PRMT3 in complex with compound 1 as well as kinetic analysis reveals an allosteric mechanism of inhibition. Mutating PRMT3 residues within the allosteric site or using compound 1 analogs that disrupt interactions with allosteric site residues both abrogated binding and inhibitory activity. These data demonstrate an allosteric mechanism for inhibition of protein arginine methyltransferases, an emerging class of therapeutic targets.


Enzyme Inhibitors/chemistry , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/chemistry , Thiadiazoles/chemistry , Urea/analogs & derivatives , Allosteric Regulation , Allosteric Site , Amino Acid Substitution , Caco-2 Cells , Catalytic Domain , Cell Membrane Permeability , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Humans , Hydrogen Bonding , Kinetics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Secondary , Protein-Arginine N-Methyltransferases/genetics , Structure-Activity Relationship , Thiadiazoles/metabolism , Urea/chemistry , Urea/metabolism
5.
J Med Chem ; 55(14): 6467-77, 2012 Jul 26.
Article En | MEDLINE | ID: mdl-22738238

Adenosine A(1) receptor (A(1)AR) agonists have antinociceptive effects in multiple preclinical models of acute and chronic pain. Although numerous A(1)AR agonists have been developed, clinical applications of these agents have been hampered by their cardiovascular side effects. Herein we report a series of novel A(1)AR agonists, some of which are structurally related to adenosine 5'-monophosphate (5'-AMP), a naturally occurring nucleotide that itself activates A(1)AR. These novel compounds potently activate A(1)AR in several orthogonal in vitro assays and are subtype selective for A(1)AR over A(2A)AR, A(2B)AR, and A(3)AR. Among them, UNC32A (3a) is orally active and has dose-dependent antinociceptive effects in wild-type mice. The antinociceptive effects of 3a were completely abolished in A(1)AR knockout mice, revealing a strict dependence on A(1)AR for activity. The apparent lack of cardiovascular side effects when administered orally and high affinity (K(i) of 36 nM for the human A(1)AR) make this compound potentially suitable as a therapeutic.


Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/pharmacology , Adenosine Monophosphate/administration & dosage , Adenosine Monophosphate/pharmacology , Analgesics/administration & dosage , Analgesics/pharmacology , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/metabolism , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Administration, Oral , Analgesics/chemistry , Analgesics/metabolism , Animals , Behavior, Animal/drug effects , HEK293 Cells , Humans , Male , Mice , Nociception/drug effects , Substrate Specificity , Temperature
6.
J Biol Chem ; 287(8): 5301-9, 2012 Feb 17.
Article En | MEDLINE | ID: mdl-22215671

Numerous receptors for ATP, ADP, and adenosine exist; however, it is currently unknown whether a receptor for the related nucleotide adenosine 5'-monophosphate (AMP) exists. Using a novel cell-based assay to visualize adenosine receptor activation in real time, we found that AMP and a non-hydrolyzable AMP analog (deoxyadenosine 5'-monophosphonate, ACP) directly activated the adenosine A(1) receptor (A(1)R). In contrast, AMP only activated the adenosine A(2B) receptor (A(2B)R) after hydrolysis to adenosine by ecto-5'-nucleotidase (NT5E, CD73) or prostatic acid phosphatase (PAP, ACPP). Adenosine and AMP were equipotent human A(1)R agonists in our real-time assay and in a cAMP accumulation assay. ACP also depressed cAMP levels in mouse cortical neurons through activation of endogenous A(1)R. Non-selective purinergic receptor antagonists (pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid and suramin) did not block adenosine- or AMP-evoked activation. Moreover, mutation of His-251 in the human A(1)R ligand binding pocket reduced AMP potency without affecting adenosine potency. In contrast, mutation of a different binding pocket residue (His-278) eliminated responses to AMP and to adenosine. Taken together, our study indicates that the physiologically relevant nucleotide AMP is a full agonist of A(1)R. In addition, our study suggests that some of the physiological effects of AMP may be direct, and not indirect through ectonucleotidases that hydrolyze this nucleotide to adenosine.


Adenosine A1 Receptor Agonists/pharmacology , Adenosine Monophosphate/pharmacology , Receptor, Adenosine A1/metabolism , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/metabolism , Animals , Cerebral Cortex/cytology , Colforsin/pharmacology , HEK293 Cells , Histidine , Humans , Hydrolysis/drug effects , Ligands , Mice , Molecular Imaging , Neurons/drug effects , Neurons/metabolism , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A2B/metabolism , Receptors, Purinergic P2Y/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis
7.
Acta Biochim Biophys Sin (Shanghai) ; 44(1): 70-9, 2012 Jan.
Article En | MEDLINE | ID: mdl-22194015

During the last decade, we saw an explosion of studies investigating the role of lysine methylation/demethylation of histones and non-histone proteins, such as p53, NF-kappaB, and E2F1. These 'Ying-Yang' post-translational modifications are important to fine-tuning the activity of these proteins. Lysine methylation and demethylation are catalyzed by protein lysine methyltransferases (PKMTs) and protein lysine demethylases (PKDMs). PKMTs, PKDMs, and their substrates have been shown to play important roles in cancers. Although the underlying mechanisms of tumorigenesis are still largely unknown, growing evidence is starting to link aberrant regulation of methylation to tumorigenesis. This review focuses on summarizing the recent progress in understanding of the function of protein lysine methylation, and in the discovery of small molecule inhibitors for PKMTs and PKDMs. We also discuss the potential and the caveats of targeting protein lysine methylation for the treatment of cancer.


Enzyme Inhibitors/therapeutic use , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Neoplasms/drug therapy , DNA-Binding Proteins/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Histocompatibility Antigens , Histone Demethylases/antagonists & inhibitors , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Methyltransferases/antagonists & inhibitors , Neoplasms/genetics , Polycomb Repressive Complex 2 , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Repressor Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
8.
Curr Chem Genomics ; 5(Suppl 1): 72-84, 2011.
Article En | MEDLINE | ID: mdl-21966347

Growing evidence suggests that protein lysine methyltransferases (PKMTs) and protein arginine methyltransferases (PRMTs) are associated with the development of various human diseases, including cancer, inflammation, and psychiatric disorders. Given the significant role of these proteins in human disease, efforts to discover selective small-molecule inhibitors of these enzymes are quickly gaining momentum. In this review, we focus on the recent progress in the discovery of selective PKMT and PRMT inhibitors. A future perspective on developing methyltransferase inhibitors is also offered.

9.
J Org Chem ; 76(7): 2094-101, 2011 Apr 01.
Article En | MEDLINE | ID: mdl-21361394

A wide variety of stabilized carbanions have been found to participate as nucleophiles in intramolecular Michael-type conjugate additions to in situ generated nitrosoalkenes to form bridged carbocyclic systems. The vinylnitroso platforms for these cyclizations have been prepared via two key steps involving ring-closing metathesis of vinyl chlorides and regioselective conversion of vinyl chlorides to α-chloroketones with sodium hypochlorite in glacial acetic acid/acetone. An alternative approach to preparation of some cyclization substrates has involved use of more reactive enol ethers as precursors to the requisite α-chloroketones. A sulfonamide anion has also been found to be an effective nucleophile in this type of reaction, leading to formation of a 6-azabicyclo[3.2.1]octane.


Anions/chemistry , Azabicyclo Compounds/chemistry , Azabicyclo Compounds/chemical synthesis , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/chemical synthesis , Cycloparaffins/chemistry , Nitroso Compounds/chemistry , Sulfonamides/chemistry , Cyclization , Magnetic Resonance Spectroscopy , Molecular Structure , Stereoisomerism
10.
Tetrahedron Lett ; 49(19): 3162-3164, 2008 May 05.
Article En | MEDLINE | ID: mdl-19421308

A wide variety of alpha-diazo-beta-ketoesters can be prepared in good overall yields via a two-step sequence involving addition of ethyl lithiodiazoacetate to aliphatic, aromatic and conjugated aldehydes followed by mild oxidation with the Dess-Martin periodinane.

12.
Bioorg Med Chem Lett ; 16(17): 4444-9, 2006 Sep 01.
Article En | MEDLINE | ID: mdl-16806925

A new series of 1,2,4-triazoles was synthesized and tested against several NNRTI-resistant HIV-1 isolates. Several of these compounds exhibited potent antiviral activities against efavirenz- and nevirapine-resistant viruses, containing K103N and/or Y181C mutations or Y188L mutation. Triazoles were first synthesized from commercially available substituted phenylthiosemicarbazides, then from isothiocyanates, and later by condensing the desired substituted anilines with thiosemicarbazones.


Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , Triazoles/chemistry , Triazoles/pharmacology , Enzyme Inhibitors/chemical synthesis , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/enzymology , Molecular Structure , Nucleosides/chemical synthesis , Nucleosides/chemistry , Nucleosides/pharmacology , Structure-Activity Relationship , Triazoles/chemical synthesis
...