Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 22(1): e3002462, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38289969

ABSTRACT

Mutations in the gene encoding Cu-Zn superoxide dismutase 1 (SOD1) cause a subset of familial amyotrophic lateral sclerosis (fALS) cases. A shared effect of these mutations is that SOD1, which is normally a stable dimer, dissociates into toxic monomers that seed toxic aggregates. Considerable research effort has been devoted to developing compounds that stabilize the dimer of fALS SOD1 variants, but unfortunately, this has not yet resulted in a treatment. We hypothesized that cyclic thiosulfinate cross-linkers, which selectively target a rare, 2 cysteine-containing motif, can stabilize fALS-causing SOD1 variants in vivo. We created a library of chemically diverse cyclic thiosulfinates and determined structure-cross-linking-activity relationships. A pre-lead compound, "S-XL6," was selected based upon its cross-linking rate and drug-like properties. Co-crystallographic structure clearly establishes the binding of S-XL6 at Cys 111 bridging the monomers and stabilizing the SOD1 dimer. Biophysical studies reveal that the degree of stabilization afforded by S-XL6 (up to 24°C) is unprecedented for fALS, and to our knowledge, for any protein target of any kinetic stabilizer. Gene silencing and protein degrading therapeutic approaches require careful dose titration to balance the benefit of diminished fALS SOD1 expression with the toxic loss-of-enzymatic function. We show that S-XL6 does not share this liability because it rescues the activity of fALS SOD1 variants. No pharmacological agent has been proven to bind to SOD1 in vivo. Here, using a fALS mouse model, we demonstrate oral bioavailability; rapid engagement of SOD1G93A by S-XL6 that increases SOD1G93A's in vivo half-life; and that S-XL6 crosses the blood-brain barrier. S-XL6 demonstrated a degree of selectivity by avoiding off-target binding to plasma proteins. Taken together, our results indicate that cyclic thiosulfinate-mediated SOD1 stabilization should receive further attention as a potential therapeutic approach for fALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Animals , Mice , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Cysteine/genetics , Mutation , Superoxide Dismutase/genetics , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics
2.
Cell Chem Biol ; 29(2): 249-258.e5, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34547225

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma low-density lipoprotein cholesterol (LDL-C) levels by promoting hepatic LDL receptor (LDLR) degradation. Therapeutic antibodies that disrupt PCSK9-LDLR binding reduce LDL-C concentrations and cardiovascular disease risk. The epidermal growth factor precursor homology domain A (EGF-A) of the LDLR serves as a primary contact with PCSK9 via a flat interface, presenting a challenge for identifying small molecule PCSK9-LDLR disruptors. We employ an affinity-based screen of 1013in vitro-translated macrocyclic peptides to identify high-affinity PCSK9 ligands that utilize a unique, induced-fit pocket and partially disrupt the PCSK9-LDLR interaction. Structure-based design led to molecules with enhanced function and pharmacokinetic properties (e.g., 13PCSK9i). In mice, 13PCSK9i reduces plasma cholesterol levels and increases hepatic LDLR density in a dose-dependent manner. 13PCSK9i functions by a unique, allosteric mechanism and is the smallest molecule identified to date with in vivo PCSK9-LDLR disruptor function.


Subject(s)
Peptides/pharmacology , Proprotein Convertase 9/metabolism , Receptors, LDL/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Peptides/chemical synthesis , Peptides/chemistry , Protein Conformation , Receptors, LDL/metabolism
3.
J Med Chem ; 64(5): 2622-2633, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33629858

ABSTRACT

Advances in the design of permeable peptides and in the synthesis of large arrays of macrocyclic peptides with diverse amino acids have evolved on parallel but independent tracks. Less precedent combines their respective attributes, thereby limiting the potential to identify permeable peptide ligands for key targets. Herein, we present novel 6-, 7-, and 8-mer cyclic peptides (MW 774-1076 g·mol-1) with passive permeability and oral exposure that feature the amino acids and thioether ring-closing common to large array formats, including DNA- and RNA-templated synthesis. Each oral peptide herein, selected from virtual libraries of partially N-methylated peptides using in silico methods, reflects the subset consistent with low energy conformations, low desolvation penalties, and passive permeability. We envision that, by retaining the backbone N-methylation pattern and consequent bias toward permeability, one can generate large peptide arrays with sufficient side chain diversity to identify permeability-biased ligands to a variety of protein targets.


Subject(s)
Peptides, Cyclic/pharmacology , Sulfides/pharmacology , Administration, Oral , Animals , Caco-2 Cells , Cell Membrane Permeability , Dogs , Humans , Madin Darby Canine Kidney Cells , Male , Methylation , Molecular Structure , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacokinetics , Protein Conformation , Rats, Sprague-Dawley , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/pharmacology , Sulfides/administration & dosage , Sulfides/chemical synthesis , Sulfides/pharmacokinetics , Thermodynamics
4.
Int J Pharm ; 585: 119519, 2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32535069

ABSTRACT

A local sustained-release drug delivery system, or depot, for intra-articular injection offers the opportunity to release a therapeutic agent directly to the joint with limited need for reinjection. A successful system would provide more consistent efficacy and minimize systemic side effects. In this paper, we explore the potential use of diclofenac, a non-steroidal anti-inflammatory drug, for use in a polymer-conjugate depot system. During the course of our exploration it was determined that "conventional ester" conjugates of diclofenac were not appropriate as upon incubation in buffer (pH 7.4) or in bovine synovial fluid, a considerable amount of undesired diclofenac-lactam was released. Thus we developed a novel linker system for diclofenac in order to minimize the production of the lactam. This new linker enables a diclofenac conjugate system with tunable release rates and minimizes the production of undesired lactam side-products.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Benzyl Alcohols/chemistry , Diclofenac/administration & dosage , Drug Delivery Systems/methods , Hydrogels/chemistry , Animals , Cattle , Chemistry, Pharmaceutical/methods , Delayed-Action Preparations , Humans , Hydrogen-Ion Concentration , Injections, Intra-Articular , Prodrugs , Synovial Fluid
5.
J Biomol Screen ; 21(6): 620-5, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26903406

ABSTRACT

A new analysis approach was evaluated for measuring plasma protein binding (PPB) of small molecules using the Agilent RapidFire high-throughput system coupled with a Sciex API 4000 mass spectrometer (RF-MS/MS). Thirty-three proprietary and 12 literature compounds were subjected to rapid equilibrium dialysis (RED) and evaluated in parallel using RF-MS/MS at 16.4 s/sample and traditional liquid chromatography-tandem mass spectrometry (LC-MS/MS) at 3.5 min/sample, thus making the RF-MS/MS analysis over 12 times faster than LC-MS/MS. The high-throughput analysis method that was developed demonstrated excellent correlation with the traditional LC-MS/MS analysis method with an r(2) value of 0.96. The RF-MS/MS analysis method was implemented to increase sample throughput, decrease turnaround time for PPB data, and decrease time burden on existing LC-MS/MS instruments.


Subject(s)
Chromatography, Liquid/methods , High-Throughput Screening Assays/methods , Proteins/chemistry , Tandem Mass Spectrometry/methods , Humans , Protein Binding , Proteins/antagonists & inhibitors , Solid Phase Extraction
6.
J Med Chem ; 56(13): 5464-72, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23738526

ABSTRACT

Glaucoma is a leading cause of vision loss and blindness, with increased intraocular pressure (IOP) a prominent risk factor. IOP can be efficaciously reduced by administration of topical agents. However, the repertoire of approved IOP-lowering drug classes is limited, and effective new alternatives are needed. Agonism of the cannabinoid receptors CB1/2 significantly reduces IOP clinically and experimentally. However, development of CB1/2 agonists has been complicated by the need to avoid cardiovascular and psychotropic side effects. 1 is a potent CB1/2 agonist that is highly excluded from the brain. In a phase I study, compound 1 eyedrops were well tolerated and generated an IOP-lowering trend but were limited in dose and exposure due to poor solubility and ocular absorption. Here we present an innovative strategy to rapidly identify compound 1 prodrugs that are efficiently metabolized to the parent compound for improved solubility and ocular permeability while maintaining low systemic exposures.


Subject(s)
Ophthalmic Solutions/pharmacology , Prodrugs/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , Area Under Curve , Eye/metabolism , Eye/physiopathology , Glaucoma/metabolism , Glaucoma/physiopathology , Glaucoma/prevention & control , Humans , Intraocular Pressure/drug effects , Male , Metabolic Clearance Rate , Models, Chemical , Molecular Structure , Ophthalmic Solutions/chemical synthesis , Ophthalmic Solutions/pharmacokinetics , Permeability , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Rats , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism
7.
ACS Med Chem Lett ; 4(6): 514-6, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-24900702

ABSTRACT

Susceptibility to metabolism is a common issue with the tert-butyl group on compounds of medicinal interest. We demonstrate an approach of removing all the fully sp(3) C-Hs from a tert-butyl group: replacing some C-Hs with C-Fs and increasing the s-character of the remaining C-Hs. This approach gave a trifluoromethylcyclopropyl group, which increased metabolic stability. Trifluoromethylcyclopropyl-containing analogues had consistently higher metabolic stability in vitro and in vivo compared to their tert-butyl-containing counterparts.

8.
J Pharm Biomed Anal ; 61: 30-7, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22177411

ABSTRACT

Unique and remarkable interferences were observed when dried blood spot (DBS) sampling was used in conjunction with liquid chromatography/mass spectrometry (LC/MS) assays. In particular, chromatographic retention time shifting and chromatographic peak shape distortion were observed, along with a severe suppression of MS signal intensity. The type of DBS cards, and chromatographic conditions were investigated using the same set of test compounds to gain insight into these interferences. It was determined that a constituent of the DBS cards, primarily sodium dodecyl sulfate (SDS), was responsible for the interferences by means of an ion-pairing mechanism. SDS formed ion pairs with compounds containing basic amine groups, which resulted in increased retention on a C(18) stationary phase, peak shape distortion and ion suppression. These interferences were greatly alleviated and/or completely overcome with non-acidic mobile phases and/or DBS cards with no SDS coating. To the best of the authors' knowledge, this is the first in-depth report of interferences induced by DBS cards.


Subject(s)
Dried Blood Spot Testing/methods , Mass Spectrometry/methods , Sodium Dodecyl Sulfate/adverse effects , Animals , Blood Specimen Collection/methods , Blood Specimen Collection/standards , Chromatography, Liquid/methods , Chromatography, Liquid/standards , Dried Blood Spot Testing/standards , Mass Spectrometry/standards , Rats , Rats, Sprague-Dawley , Sodium Dodecyl Sulfate/analysis
9.
Proc Natl Acad Sci U S A ; 108(17): 6739-44, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21502533

ABSTRACT

The search for novel therapeutic interventions for viral disease is a challenging pursuit, hallmarked by the paucity of antiviral agents currently prescribed. Targeting of viral proteins has the inextricable challenge of rise of resistance. Safe and effective vaccines are not possible for many viral pathogens. New approaches are required to address the unmet medical need in this area. We undertook a cell-based high-throughput screen to identify leads for development of drugs to treat respiratory syncytial virus (RSV), a serious pediatric pathogen. We identified compounds that are potent (nanomolar) inhibitors of RSV in vitro in HEp-2 cells and in primary human bronchial epithelial cells and were shown to act postentry. Interestingly, two scaffolds exhibited broad-spectrum activity among multiple RNA viruses. Using the chemical matter as a probe, we identified the targets and identified a common cellular pathway: the de novo pyrimidine biosynthesis pathway. Both targets were validated in vitro and showed no significant cell cytotoxicity except for activity against proliferative B- and T-type lymphoid cells. Corollary to this finding was to understand the consequences of inhibition of the target to the host. An in vivo assessment for antiviral efficacy failed to demonstrate reduced viral load, but revealed microscopic changes and a trend toward reduced pyrimidine pools and findings in histopathology. We present here a discovery program that includes screen, target identification, validation, and druggability that can be broadly applied to identify and interrogate other host factors for antiviral effect starting from chemical matter of unknown target/mechanism of action.


Subject(s)
Antiviral Agents , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/metabolism , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cell Proliferation/drug effects , Chlorocebus aethiops , Dogs , Dose-Response Relationship, Drug , HeLa Cells , Humans , Jurkat Cells , Respiratory Syncytial Virus Infections/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...