Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Med Chem ; 67(3): 2220-2235, 2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-38284169

RÉSUMÉ

Thymic stromal lymphopoietin (TSLP) is an epithelial-derived pro-inflammatory cytokine involved in the development of asthma and other atopic diseases. We used Bicycle Therapeutics' proprietary phage display platform to identify bicyclic peptides (Bicycles) with high affinity for TSLP, a target that is difficult to drug with conventional small molecules due to the extended protein-protein interactions it forms with both receptors. The hit series was shown to bind to TSLP in a hotspot, that is also used by IL-7Rα. Guided by the first X-ray crystal structure of a small peptide binding to TSLP and the identification of key metabolites, we were able to improve the proteolytic stability of this series in lung S9 fractions without sacrificing binding affinity. This resulted in the potent Bicycle 46 with nanomolar affinity to TSLP (KD = 13 nM), low plasma clearance of 6.4 mL/min/kg, and an effective half-life of 46 min after intravenous dosing to rats.


Sujet(s)
Asthme , Lymphopoïétine stromale thymique , Animaux , Rats , Asthme/traitement médicamenteux , Cyclisme , Cytokines/métabolisme , Peptides cycliques/composition chimique , Peptides cycliques/métabolisme
2.
Nat Commun ; 14(1): 3583, 2023 06 16.
Article de Anglais | MEDLINE | ID: mdl-37328472

RÉSUMÉ

COVID-19 has stimulated the rapid development of new antibody and small molecule therapeutics to inhibit SARS-CoV-2 infection. Here we describe a third antiviral modality that combines the drug-like advantages of both. Bicycles are entropically constrained peptides stabilized by a central chemical scaffold into a bi-cyclic structure. Rapid screening of diverse bacteriophage libraries against SARS-CoV-2 Spike yielded unique Bicycle binders across the entire protein. Exploiting Bicycles' inherent chemical combinability, we converted early micromolar hits into nanomolar viral inhibitors through simple multimerization. We also show how combining Bicycles against different epitopes into a single biparatopic agent allows Spike from diverse variants of concern (VoC) to be targeted (Alpha, Beta, Delta and Omicron). Finally, we demonstrate in both male hACE2-transgenic mice and Syrian golden hamsters that both multimerized and biparatopic Bicycles reduce viraemia and prevent host inflammation. These results introduce Bicycles as a potential antiviral modality to tackle new and rapidly evolving viruses.


Sujet(s)
COVID-19 , SARS-CoV-2 , Mâle , Animaux , Cricetinae , Souris , Antiviraux/pharmacologie , Peptides/pharmacologie , Anticorps , Mesocricetus , Souris transgéniques , Glycoprotéine de spicule des coronavirus/génétique
3.
J Med Chem ; 65(21): 14337-14347, 2022 11 10.
Article de Anglais | MEDLINE | ID: mdl-36204777

RÉSUMÉ

Bicycle toxin conjugates (BTCs) are a promising new class of molecules for targeted delivery of toxin payloads into tumors. Herein we describe the discovery of BT8009, a Nectin-4 targeting BTC currently under clinical evaluation. Nectin-4 is overexpressed in multiple tumor types and is a clinically validated target for selective delivery of cytotoxic payloads. A Nectin-4 targeting bicyclic peptide was identified by phage display, which showed highly selective binding for Nectin-4 but suffered from low plasma stability and poor physicochemical properties. Multiparameter chemical optimization involving introduction of non-natural amino acids resulted in a lead Bicycle that demonstrated high affinity for Nectin-4, good stability in biological matrices, and a much-improved physicochemical profile. The optimized Bicycle was conjugated to the cytotoxin Monomethyl auristatin E via a cleavable linker to give the targeted drug conjugate BT8009, which demonstrates potent anticancer activity in in vivo rodent models.


Sujet(s)
Antinéoplasiques , Immunoconjugués , Immunotoxines , Tumeurs , Humains , Nectines , Cyclisme , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/composition chimique , Molécules d'adhérence cellulaire , Lignée cellulaire tumorale
4.
Mol Cancer Ther ; 21(12): 1747-1756, 2022 12 02.
Article de Anglais | MEDLINE | ID: mdl-36112771

RÉSUMÉ

Multiple tumor types overexpress Nectin-4 and the antibody-drug conjugate (ADC), enfortumab vedotin (EV) shows striking efficacy in clinical trials for metastatic urothelial cancer, which expresses high levels of Nectin-4, validating Nectin-4 as a clinical target for toxin delivery in this indication. Despite excellent data in urothelial cancer, little efficacy data are reported for EV in other Nectin-4 expressing tumors and EV therapy can produce significant toxicities in many patients, frequently leading to discontinuation of treatment. Thus, additional approaches to this target with the potential to extend utility and reduce toxicity are warranted. We describe the preclinical development of BT8009, a "Bicycle Toxin Conjugate" (BTC) consisting of a Nectin-4-binding bicyclic peptide, a cleavable linker system and the cell penetrant toxin mono-methylauristatin E (MMAE). BT8009 shows significant antitumor activity in preclinical tumor models, across a variety of cancer indications and is well tolerated in preclinical safety studies. In several models, it shows superior or equivalent antitumor activity to an EV analog. As a small hydrophilic peptide-based drug BT8009 rapidly diffuses from the systemic circulation, through tissues to penetrate the tumor and target tumor cells. It is renally eliminated from the circulation, with a half-life of 1-2 hours in rat and non-human primate. These physical and PK characteristics differentiate BT8009 from ADCs and may provide benefit in terms of tumor penetration and reduced systemic exposure. BT8009 is currently in a Phase 1/2 multicenter clinical trial across the US, Canada, and Europe, enrolling patients with advanced solid tumors associated with Nectin-4 expression.


Sujet(s)
Carcinome transitionnel , Immunoconjugués , Immunotoxines , Rats , Animaux , Nectines , Cyclisme , Immunoconjugués/pharmacologie , Immunoconjugués/usage thérapeutique , Molécules d'adhérence cellulaire/métabolisme , Carcinome transitionnel/traitement médicamenteux
5.
J Immunother Cancer ; 9(1)2021 01.
Article de Anglais | MEDLINE | ID: mdl-33500260

RÉSUMÉ

BACKGROUND: In contrast to immune checkpoint inhibitors, the use of antibodies as agonists of immune costimulatory receptors as cancer therapeutics has largely failed. We sought to address this problem using a new class of modular synthetic drugs, termed tumor-targeted immune cell agonists (TICAs), based on constrained bicyclic peptides (Bicycles). METHODS: Phage libraries displaying Bicycles were panned for binders against tumor necrosis factor (TNF) superfamily receptors CD137 and OX40, and tumor antigens EphA2, Nectin-4 and programmed death ligand 1. The CD137 and OX40 Bicycles were chemically conjugated to tumor antigen Bicycles with different linkers and stoichiometric ratios of binders to obtain a library of low molecular weight TICAs (MW <8 kDa). The TICAs were evaluated in a suite of in vitro and in vivo assays to characterize their pharmacology and mechanism of action. RESULTS: Linking Bicycles against costimulatory receptors (e.g., CD137) to Bicycles against tumor antigens (e.g., EphA2) created potent agonists that activated the receptors selectively in the presence of tumor cells expressing these antigens. An EphA2/CD137 TICA (BCY12491) efficiently costimulated human peripheral blood mononuclear cells in vitro in the presence of EphA2 expressing tumor cell lines as measured by the increased secretion of interferon γ and interleukin-2. Treatment of C57/Bl6 mice transgenic for the human CD137 extracellular domain (huCD137) bearing EphA2-expressing MC38 tumors with BCY12491 resulted in the infiltration of CD8+ T cells, elimination of tumors and generation of immunological memory. BCY12491 was cleared quickly from the circulation (plasma t1/2 in mice of 1-2 hr), yet intermittent dosing proved effective. CONCLUSION: Tumor target-dependent CD137 agonism using a novel chemical approach (TICAs) afforded elimination of tumors with only intermittent dosing suggesting potential for a wide therapeutic index in humans. This work unlocks a new path to effective cancer immunotherapy via agonism of TNF superfamily receptors.


Sujet(s)
Tumeurs/traitement médicamenteux , Peptides cycliques/administration et posologie , Récepteur EphA2/agonistes , Antigènes CD137/agonistes , Cellules A549 , Animaux , Antigènes néoplasiques/métabolisme , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Femelle , Cellules HT29 , Humains , Cellules Jurkat , Souris , Souris transgéniques , Tumeurs/génétique , Tumeurs/immunologie , Cellules PC-3 , Banque de peptides , Peptides cycliques/composition chimique , Peptides cycliques/pharmacologie , Récepteur au OX40/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
6.
ACS Infect Dis ; 6(9): 2355-2361, 2020 09 11.
Article de Anglais | MEDLINE | ID: mdl-32697574

RÉSUMÉ

The treatment of infection by Gram-negative bacteria is increasingly challenging as resistance to existing antibiotics spreads. Constrained peptides, selected for high target specificity and affinity via library display technologies, are an emerging therapeutic modality in many disease areas and may be a fertile source of new antibiotics. Currently, the utility of constrained peptides and other large molecules as antibiotics is limited by the outer membrane (OM) barrier of Gram-negative bacteria. However, the addition of certain moieties to large molecules can confer the ability to cross the OM; these moieties function as intramolecular trans-OM "vectors". Here, we present a method to systematically assess the carrying capacity of candidate trans-OM vectors using a real-time luminescence assay ("SLALOM", Split Luciferase Assay for Live monitoring of Outer Membrane transit), reporting on periplasmic entry. We demonstrate the usefulness of our tools by constructing a 3800 Da chimeric compound composed of a constrained bicyclic peptide (Bicycle) with a periplasmic target, linked to an intramolecular peptide vector; the resulting chimera is a broad-spectrum inhibitor of pathogenic Gram-negative bacterial growth.


Sujet(s)
Bactéries à Gram négatif , Périplasme , Antibactériens/pharmacologie , Chimère
7.
Mol Cancer Ther ; 19(7): 1385-1394, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32398269

RÉSUMÉ

The EphA2 receptor is found at high levels in tumors and low levels in normal tissue and high EphA2 expression in biopsies is a predictor of poor outcome in patients. Drug discovery groups have therefore sought to develop EphA2-based therapies using small molecule, peptide, and nanoparticle-based approaches (1-3). However, until now only EphA2-targeting antibody-drug conjugates (ADC) have entered clinical development. For example, MEDI-547 is an EphA2-targeting ADC that displayed encouraging antitumor activity in preclinical models and progressed to phase I clinical testing in man. Here we describe the development of BT5528, a bicyclic peptide ("Bicycle") conjugated to the auristatin derivative maleimidocaproyl-monomethyl auristatin E to generate the Bicycle toxin conjugate BT5528. The report compares and contrasts the Pharmacokinetics (PK) characteristics of antibody and Bicycle-based targeting systems and discusses how the PK and payload characteristics of different delivery systems impact the efficacy-toxicity trade off which is key to the development of successful cancer therapies. We show that BT5528 gives rise to rapid update into tumors and fast renal elimination followed by persistent toxin levels in tumors without prolonged exposure of parent drug in the vasculature. This fast in, fast out kinetics gave rise to more favorable toxicology findings in rats and monkeys than were observed with MEDI-547 in preclinical and clinical studies.Graphical Abstract: http://mct.aacrjournals.org/content/molcanther/19/7/1385/F1.large.jpg.


Sujet(s)
Antinéoplasiques/pharmacologie , Systèmes de délivrance de médicaments , Fibrosarcome/traitement médicamenteux , Oligopeptides/composition chimique , Peptides cycliques/pharmacologie , Récepteur EphA2/métabolisme , Animaux , Anticorps monoclonaux/pharmacologie , Antinéoplasiques/pharmacocinétique , Apoptose , Prolifération cellulaire , Femelle , Fibrosarcome/métabolisme , Fibrosarcome/anatomopathologie , Humains , Immunotoxines/pharmacocinétique , Immunotoxines/pharmacologie , Mâle , Souris , Souris de lignée BALB C , Souris nude , Oligopeptides/administration et posologie , Peptides cycliques/pharmacocinétique , Récepteur EphA2/génétique , Distribution tissulaire , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
8.
J Med Chem ; 63(8): 4107-4116, 2020 04 23.
Article de Anglais | MEDLINE | ID: mdl-32202781

RÉSUMÉ

Bicycles are constrained bicyclic peptides that represent a promising binding modality for use in targeted drug conjugates. A phage display screen against EphA2, a receptor tyrosine kinase highly expressed in a number of solid tumors, identified a number of Bicycle families with low nanomolar affinity. A Bicycle toxin conjugate (BTC) was generated by derivatization of one of these Bicycles with the potent cytotoxin DM1 via a cleavable linker. This BTC demonstrated potent antitumor activity in vivo but was poorly tolerated, which was hypothesized to be the result of undesired liver uptake caused by poor physicochemical properties. Chemical optimization of a second Bicycle, guided by structural biology, provided a high affinity, metabolically stable Bicycle with improved physicochemical properties. A BTC incorporating this Bicycle also demonstrated potent antitumor activity and was very well tolerated when compared to the initial BTC. Phage display selection followed by chemical optimization of Bicycles can deliver potent drug conjugates with favorable pharmaceutical properties.


Sujet(s)
Composés hétérocycliques bicycliques/administration et posologie , Cytotoxines/administration et posologie , Systèmes de délivrance de médicaments/méthodes , Éphrine A2/antagonistes et inhibiteurs , Séquence d'acides aminés , Animaux , Composés hétérocycliques bicycliques/composition chimique , Composés hétérocycliques bicycliques/métabolisme , Cytotoxines/composition chimique , Cytotoxines/métabolisme , Éphrine A2/métabolisme , Femelle , Humains , Foie/imagerie diagnostique , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Mâle , Souris , Souris de lignée BALB C , Souris nude , Structure secondaire des protéines , Structure tertiaire des protéines , Récepteur EphA2 , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
9.
Cancer Res ; 79(4): 841-852, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-30606721

RÉSUMÉ

Molecular imaging of cancers using probes specific for tumor-associated target proteins offers a powerful solution for providing information regarding selection of targeted therapy, patient stratification, and response to therapy. Here we demonstrate the power of bicyclic peptides as targeting probes, exemplified with the tumor-overexpressed matrix metalloproteinase MT1-MMP as a target. A bicyclic peptide with subnanomolar affinity towards MT1-MMP was identified, and its radioconjugate showed selective tumor uptake in an HT1080 xenograft mouse model. Proteolytic stabilization of the peptide by chemical modification significantly enhanced the in vivo tumor signal [from 2.5%ID/g to 12%ID/g at 1 hour post injection (p.i.)]. Studies using mouse xenograft models with different cell lines show a robust correlation between tumor signals and in vivo MT1-MMP expression levels. Fatty acid modification of the bicyclic peptide extended its circulating half-life, resulting in increased tumor signals (36%ID/g at 6 hours p.i.). Comparative work with an equipotent radiolabeled MT1-MMP targeting antibody demonstrated starkly differential biodistribution and tumor accumulation properties, with the tumor signal slowly increasing to 6.2%ID/g within 48 hours. The rapid tumor penetration characteristics of bicyclic peptides, coupled with high potency and chemical versatility, thus offer high-contrast imaging probes for clinical diagnostics with compelling additional potential in targeted therapy.Significance: This work demonstrates the potential of bicyclic peptides as a platform for the development of high-contrast imaging probes for potential use in clinical cancer diagnostics and molecularly targeted therapeutics.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Antienzymes/pharmacologie , Fibrosarcome/traitement médicamenteux , Fibrosarcome/métabolisme , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Matrix metalloproteinase 14/composition chimique , Peptides cycliques/pharmacologie , Animaux , Anticorps monoclonaux/pharmacocinétique , Apoptose , Prolifération cellulaire , Antienzymes/pharmacocinétique , Fibrosarcome/imagerie diagnostique , Fibrosarcome/anatomopathologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Traitement d'image par ordinateur/méthodes , Mâle , Matrix metalloproteinase 14/métabolisme , Souris , Souris de lignée BALB C , Souris nude , Peptides cycliques/pharmacocinétique , Tomographie par émission de positons , Distribution tissulaire , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
10.
J Med Chem ; 61(7): 2823-2836, 2018 04 12.
Article de Anglais | MEDLINE | ID: mdl-29517911

RÉSUMÉ

Plasma kallikrein, a member of the kallikrein-kinin system, catalyzes the release of the bioactive peptide bradykinin, which induces inflammation, vasodilation, vessel permeability, and pain. Preclinical evidence implicates the activity of plasma kallikrein in diabetic retinopathy, which is a leading cause of visual loss in patients suffering from diabetes mellitus. Employing a technology based on phage-display combined with chemical cyclization, we have identified highly selective bicyclic peptide inhibitors with nano- and picomolar potencies toward plasma kallikrein. Stability in biological matrices was either intrinsic to the peptide or engineered via the introduction of non-natural amino acids and nonpeptidic bonds. The peptides prevented bradykinin release in vitro, and in vivo efficacy was demonstrated in both a rat paw edema model and in rodent models of diabetes-induced retinal permeability. With a highly extended half-life of ∼40 h in rabbit eyes following intravitreal administration, the bicyclic peptides are promising novel agents for the treatment of diabetic retinopathy and diabetic macular edema.


Sujet(s)
Composés bicycliques pontés/synthèse chimique , Composés bicycliques pontés/pharmacologie , Complications du diabète/traitement médicamenteux , Rétinopathie diabétique/traitement médicamenteux , Oedème maculaire/traitement médicamenteux , Oedème maculaire/étiologie , Kallicréine plasmatique/antagonistes et inhibiteurs , Inhibiteurs de protéases/synthèse chimique , Inhibiteurs de protéases/pharmacologie , Animaux , Bradykinine/métabolisme , Oedème/traitement médicamenteux , Oeil/métabolisme , Pied/anatomopathologie , Période , Injections intravitréennes , Mâle , Souris , Souris de lignée C57BL , Perméabilité , Inhibiteurs de protéases/administration et posologie , Lapins , Rats , Rat Sprague-Dawley , Relation structure-activité , Spécificité du substrat , Corps vitré/composition chimique , Corps vitré/métabolisme
11.
PLoS One ; 8(1): e52179, 2013.
Article de Anglais | MEDLINE | ID: mdl-23300965

RÉSUMÉ

Salicylidene acylhydrazides identified as inhibitors of virulence-mediating type III secretion systems (T3SSs) potentially target their inner membrane export apparatus. They also lead to inhibition of flagellar T3SS-mediated swimming motility in Salmonella enterica serovar. Typhimurium. We show that INP0404 and INP0405 act by reducing the number of flagella/cell. These molecules still inhibit motility of a Salmonella ΔfliH-fliI-fliJ/flhB((P28T)) strain, which lacks three soluble components of the flagellar T3S apparatus, suggesting that they are not the target of this drug family. We implemented a genetic screen to search for the inhibitors' molecular target(s) using motility assays in the ΔfliH-fliI/flhB((P28T)) background. Both mutants identified were more motile than the background strain in the absence of the drugs, although HM18 was considerably more so. HM18 was more motile than its parent strain in the presence of both drugs while DI15 was only insensitive to INP0405. HM18 was hypermotile due to hyperflagellation, whereas DI15 was not hyperflagellated. HM18 was also resistant to a growth defect induced by high concentrations of the drugs. Whole-genome resequencing of HM18 indicated two alterations within protein coding regions, including one within atpB, which encodes the inner membrane a-subunit of the F(O)F(1)-ATP synthase. Reverse genetics indicated that the alteration in atpB was responsible for all of HM18's phenotypes. Genome sequencing of DI15 uncovered a single A562P mutation within a gene encoding the flagellar inner membrane protein FlhA, the direct role of which in mediating drug insensitivity could not be confirmed. We discuss the implications of these findings in terms of T3SS export apparatus function and drug target identification.


Sujet(s)
Antibactériens/pharmacologie , Flagelles/métabolisme , Hydrazines/pharmacologie , Acide salicylique/pharmacologie , Salmonella enterica/effets des médicaments et des substances chimiques , Salmonella enterica/génétique , Allèles , Protéines bactériennes/métabolisme , Chromosomes/ultrastructure , Résistance bactérienne aux médicaments/génétique , Flagelles/effets des médicaments et des substances chimiques , Délétion de gène , Mouvement , Mutation , Plasmides/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...