Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nature ; 622(7982): 393-401, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37821590

RÉSUMÉ

Recent human decedent model studies1,2 and compassionate xenograft use3 have explored the promise of porcine organs for human transplantation. To proceed to human studies, a clinically ready porcine donor must be engineered and its xenograft successfully tested in nonhuman primates. Here we describe the design, creation and long-term life-supporting function of kidney grafts from a genetically engineered porcine donor transplanted into a cynomolgus monkey model. The porcine donor was engineered to carry 69 genomic edits, eliminating glycan antigens, overexpressing human transgenes and inactivating porcine endogenous retroviruses. In vitro functional analyses showed that the edited kidney endothelial cells modulated inflammation to an extent that was indistinguishable from that of human endothelial cells, suggesting that these edited cells acquired a high level of human immune compatibility. When transplanted into cynomolgus monkeys, the kidneys with three glycan antigen knockouts alone experienced poor graft survival, whereas those with glycan antigen knockouts and human transgene expression demonstrated significantly longer survival time, suggesting the benefit of human transgene expression in vivo. These results show that preclinical studies of renal xenotransplantation could be successfully conducted in nonhuman primates and bring us closer to clinical trials of genetically engineered porcine renal grafts.


Sujet(s)
Rejet du greffon , Transplantation rénale , Macaca fascicularis , Suidae , Transplantation hétérologue , Animaux , Humains , Animal génétiquement modifié , Cellules endothéliales/immunologie , Cellules endothéliales/métabolisme , Rejet du greffon/immunologie , Rejet du greffon/prévention et contrôle , Transplantation rénale/méthodes , Polyosides/déficit , Suidae/génétique , Transplantation hétérologue/méthodes , Transgènes/génétique
2.
J Control Release ; 303: 34-41, 2019 06 10.
Article de Anglais | MEDLINE | ID: mdl-30928488

RÉSUMÉ

Adherence to medication regimens is a major barrier to effective treatment in many disease areas, notably in dementia which causes cognitive impairment that reduces patients' awareness of non-adherence and their ability to manage medication. The development of oral dosage forms that can be infrequently dosed, and therefore improve adherence rate and facilitate direct observed therapy, has been a goal for decades. We describe the first demonstration of an oral formulation that achieves >7-day gastric retention and sustained pharmacokinetics in the challenging dog model. Gastric retention requires physical resistance of the dosage form to gastric emptying forces, which are known to be stronger in dogs than in humans, making successful gastric retention in dogs a stringent test for predicting human translatability. This formulation of memantine hydrochloride is the first oral dosage form that achieves multi-day drug release with near zero-order kinetics and efficient delivery. In the dog model, relative memantine bioavailability approaches 100% with sustained plasma levels of memantine over seven days and profiles that can be tuned by varying components of the formulation. A single gastric resident dosage form achieves an AUC equivalent to 7 daily treatments with the marketed daily capsule, with a Cmax that is no higher than the daily product. PK modeling predicts that the gastroretentive formulation will maintain therapeutic blood levels in humans when administered once weekly. The formulation methodology presented here is applicable to many water soluble drugs and may enable the development of long-acting oral therapies for a wide variety of conditions.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Antagonistes des acides aminés excitateurs/administration et posologie , Mémantine/administration et posologie , Administration par voie orale , Animaux , Biodisponibilité , Préparations à action retardée/administration et posologie , Préparations à action retardée/pharmacocinétique , Chiens , Calendrier d'administration des médicaments , Antagonistes des acides aminés excitateurs/sang , Antagonistes des acides aminés excitateurs/pharmacocinétique , Interactions aliments-médicaments , Humains , Mâle , Adhésion au traitement médicamenteux , Mémantine/sang , Mémantine/pharmacocinétique , Modèles biologiques , Récepteurs du N-méthyl-D-aspartate/antagonistes et inhibiteurs
3.
ACS Med Chem Lett ; 8(2): 196-200, 2017 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-28197311

RÉSUMÉ

Several monoclonal antibodies and inhibitors targeting CD38, an ectoenzyme overexpressed on malignant plasma cells, have previously been discovered. Herein, we expand structure-activity relationships of reported small-molecule thiazoloquinolinones and show that several 4-cyclohexylamino analogues have potent binding affinity for CD38 using surface plasmon resonance. Moreover, active amine analogues could be acylated and functionalized with alkyne and fluorescein groups. Fluorescein analogue 21 bound selectively to CD38 overexpressing cells, demonstrating the potential utility of thiazoloquinolinones as small-molecule conjugates for the delivery of therapeutic and imaging agents.

4.
J Control Release ; 229: 106-119, 2016 05 10.
Article de Anglais | MEDLINE | ID: mdl-27001894

RÉSUMÉ

The present studies were aimed at formulating AZD2811-loaded polylactic acid-polyethylene glycol (PLA-PEG) nanoparticles with adjustable release rates without altering the chemical structures of the polymer or active pharmaceutical ingredient (API). This was accomplished through the use of a hydrophobic ion pairing approach. A series of AZD2811-containing nanoparticles with a variety of hydrophobic counterions including oleic acid, 1-hydroxy-2-naphthoic acid, cholic acid, deoxycholic acid, dioctylsulfosuccinic acid, and pamoic acid is described. The hydrophobicity of AZD2811 was increased through formation of ion pairs with these hydrophobic counterions, producing nanoparticles with exceptionally high drug loading-up to five fold higher encapsulation efficiency and drug loading compared to nanoparticles made without hydrophobic ion pairs. Furthermore, the rate at which the drug was released from the nanoparticles could be controlled by employing counterions with various hydrophobicities and structures, resulting in release half-lives ranging from about 2 to 120h using the same polymer, nanoparticle size, and nanoemulsion process. Process recipe variables affecting drug load and release rate were identified, including pH and molarity of quench buffer. Ion pair formation between AZD2811 and pamoic acid as a model counterion was investigated using solubility enhancement as well as nuclear magnetic resonance spectroscopy to demonstrate solution-state interactions. Further evidence for an ion pairing mechanism of controlled release was provided through the measurement of API and counterion release profiles using high-performance liquid chromatography, which had stoichiometric relationships. Finally, Raman spectra of an AZD2811-pamoate salt compared well with those of the formulated nanoparticles, while single components (AZD2811, pamoic acid) alone did not. A library of AZD2811 batches was created for analytical and preclinical characterization. Dramatically improved preclinical efficacy and tolerability data were generated for the pamoic acid lead formulation, which has been selected for evaluation in a Phase 1 clinical trial (ClinicalTrials.gov Identifier NCT 02579226). This work clearly demonstrates the importance of assessing a wide range of drug release rates during formulation screening as a critical step for new drug product development, and how utilizing hydrophobic ion pairing enabled this promising nanoparticle formulation to proceed into clinical development.


Sujet(s)
Acétanilides/administration et posologie , Antinéoplasiques , Systèmes de délivrance de médicaments , Nanoparticules , Organophosphates , Promédicaments , Quinazolines/administration et posologie , Acétanilides/composition chimique , Acétanilides/pharmacocinétique , Acétanilides/usage thérapeutique , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/usage thérapeutique , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/anatomopathologie , Lignée cellulaire tumorale , Acide cholique/composition chimique , Acide désoxycholique/composition chimique , Sulfo-succinate de dioctyle/composition chimique , Humains , Interactions hydrophobes et hydrophiles , Mâle , Souris nude , Nanoparticules/administration et posologie , Nanoparticules/composition chimique , Nanoparticules/usage thérapeutique , Naphtols/composition chimique , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Organophosphates/administration et posologie , Organophosphates/composition chimique , Organophosphates/pharmacocinétique , Organophosphates/usage thérapeutique , Polyéthylène glycols/composition chimique , Promédicaments/administration et posologie , Promédicaments/composition chimique , Promédicaments/pharmacocinétique , Promédicaments/usage thérapeutique , Quinazolines/composition chimique , Quinazolines/pharmacocinétique , Quinazolines/usage thérapeutique , Rat nude , Rat Wistar , Charge tumorale/effets des médicaments et des substances chimiques
5.
Sci Transl Med ; 8(325): 325ra17, 2016 Feb 10.
Article de Anglais | MEDLINE | ID: mdl-26865565

RÉSUMÉ

Efforts to apply nanotechnology in cancer have focused almost exclusively on the delivery of cytotoxic drugs to improve therapeutic index. There has been little consideration of molecularly targeted agents, in particular kinase inhibitors, which can also present considerable therapeutic index limitations. We describe the development of Accurin polymeric nanoparticles that encapsulate the clinical candidate AZD2811, an Aurora B kinase inhibitor, using an ion pairing approach. Accurins increase biodistribution to tumor sites and provide extended release of encapsulated drug payloads. AZD2811 nanoparticles containing pharmaceutically acceptable organic acids as ion pairing agents displayed continuous drug release for more than 1 week in vitro and a corresponding extended pharmacodynamic reduction of tumor phosphorylated histone H3 levels in vivo for up to 96 hours after a single administration. A specific AZD2811 nanoparticle formulation profile showed accumulation and retention in tumors with minimal impact on bone marrow pathology, and resulted in lower toxicity and increased efficacy in multiple tumor models at half the dose intensity of AZD1152, a water-soluble prodrug of AZD2811. These studies demonstrate that AZD2811 can be formulated in nanoparticles using ion pairing agents to give improved efficacy and tolerability in preclinical models with less frequent dosing. Accurins specifically, and nanotechnology in general, can increase the therapeutic index of molecularly targeted agents, including kinase inhibitors targeting cell cycle and oncogenic signal transduction pathways, which have to date proved toxic in humans.


Sujet(s)
Aurora kinases/antagonistes et inhibiteurs , Nanoparticules/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Animaux , Aurora kinases/métabolisme , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/anatomopathologie , Lignée cellulaire tumorale , Libération de médicament , Femelle , Humains , Mâle , Spectrométrie de masse , Souris , Souris SCID , Organophosphates/composition chimique , Organophosphates/pharmacocinétique , Organophosphates/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacocinétique , Quinazolines/composition chimique , Quinazolines/pharmacocinétique , Quinazolines/pharmacologie , Rat nude , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Clin Cancer Res ; 22(13): 3157-63, 2016 07 01.
Article de Anglais | MEDLINE | ID: mdl-26847057

RÉSUMÉ

PURPOSE: First-in-human phase I trial to determine the safety, pharmacokinetics, and antitumor activity of BIND-014, a novel, tumor prostate-specific membrane antigen (PSMA)-targeted nanoparticle, containing docetaxel. EXPERIMENTAL DESIGN: Patients with advanced solid tumors received BIND-014 every three weeks (n = 28) or weekly (n = 27), with dose levels ranging from 3.5 to 75 mg/m(2) and 15 to 45 mg/m(2), respectively. RESULTS: BIND-014 was generally well tolerated, with no unexpected toxicities. The most common drug-related toxicities (>20% of patients) on either schedule included neutropenia, fatigue, anemia, alopecia, and diarrhea. BIND-014 demonstrated a dose-linear pharmacokinetic profile, distinct from docetaxel, with prolonged persistence of docetaxel-encapsulated circulating nanoparticles. Of the 52 patients evaluable for response, one had a complete response (cervical cancer on the every three week schedule) and five had partial responses (ampullary adenocarcinoma, non-small cell lung, and prostate cancers on the every-three-week schedule, and breast and gastroesophageal cancers on the weekly schedule). Responses were noted in both PSMA-detectable and -undetectable tumors. CONCLUSIONS: BIND-014 was generally well tolerated, with predictable and manageable toxicity and a unique pharmacokinetic profile compared with conventional docetaxel. Clinical activity was noted in multiple tumor types. The recommended phase II dose of BIND-014 is 60 mg/m(2) every three weeks or 40 mg/m(2) weekly. Clin Cancer Res; 22(13); 3157-63. ©2016 AACR.


Sujet(s)
Vecteurs de médicaments/pharmacocinétique , Vecteurs de médicaments/usage thérapeutique , Glutamate carboxypeptidase II/antagonistes et inhibiteurs , Nanoparticules/usage thérapeutique , Tumeurs/traitement médicamenteux , Taxoïdes/usage thérapeutique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes de surface , Docetaxel , Calendrier d'administration des médicaments , Vecteurs de médicaments/effets indésirables , Femelle , Humains , Mâle , Adulte d'âge moyen , Nanoparticules/effets indésirables , Tumeurs/anatomopathologie
7.
Sci Transl Med ; 4(128): 128ra39, 2012 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-22491949

RÉSUMÉ

We describe the development and clinical translation of a targeted polymeric nanoparticle (TNP) containing the chemotherapeutic docetaxel (DTXL) for the treatment of patients with solid tumors. DTXL-TNP is targeted to prostate-specific membrane antigen, a clinically validated tumor antigen expressed on prostate cancer cells and on the neovasculature of most nonprostate solid tumors. DTXL-TNP was developed from a combinatorial library of more than 100 TNP formulations varying with respect to particle size, targeting ligand density, surface hydrophilicity, drug loading, and drug release properties. Pharmacokinetic and tissue distribution studies in rats showed that the NPs had a blood circulation half-life of about 20 hours and minimal liver accumulation. In tumor-bearing mice, DTXL-TNP exhibited markedly enhanced tumor accumulation at 12 hours and prolonged tumor growth suppression compared to a solvent-based DTXL formulation (sb-DTXL). In tumor-bearing mice, rats, and nonhuman primates, DTXL-TNP displayed pharmacokinetic characteristics consistent with prolonged circulation of NPs in the vascular compartment and controlled release of DTXL, with total DTXL plasma concentrations remaining at least 100-fold higher than sb-DTXL for more than 24 hours. Finally, initial clinical data in patients with advanced solid tumors indicated that DTXL-TNP displays a pharmacological profile differentiated from sb-DTXL, including pharmacokinetics characteristics consistent with preclinical data and cases of tumor shrinkage at doses below the sb-DTXL dose typically used in the clinic.


Sujet(s)
Antigènes de surface/métabolisme , Glutamate carboxypeptidase II/métabolisme , Nanoparticules/composition chimique , Taxoïdes/pharmacologie , Taxoïdes/pharmacocinétique , Animaux , Lignée cellulaire tumorale , Docetaxel , Humains , Mâle , Souris , Nanoparticules/administration et posologie , Polymères/composition chimique , Rats , Taxoïdes/administration et posologie , Taxoïdes/composition chimique , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Blood ; 119(13): 3024-30, 2012 Mar 29.
Article de Anglais | MEDLINE | ID: mdl-22246033

RÉSUMÉ

Despite proven benefits, prophylactic treatment for hemophilia A is hampered by the short half-life of factor VIII. A recombinant factor VIII-Fc fusion protein (rFVIIIFc) was constructed to determine the potential for reduced frequency of dosing. rFVIIIFc has an ∼ 2-fold longer half-life than rFVIII in hemophilia A (HemA) mice and dogs. The extension of rFVIIIFc half-life requires interaction of Fc with the neonatal Fc receptor (FcRn). In FcRn knockout mice, the extension of rFVIIIFc half-life is abrogated, and is restored in human FcRn transgenic mice. The Fc fusion has no impact on FVIII-specific activity. rFVIIIFc has comparable acute efficacy as rFVIII in treating tail clip injury in HemA mice, and fully corrects whole blood clotting time (WBCT) in HemA dogs immediately after dosing. Furthermore, consistent with prolonged half-life, rFVIIIFc shows 2-fold longer prophylactic efficacy in protecting HemA mice from tail vein transection bleeding induced 24-48 hours after dosing. In HemA dogs, rFVIIIFc also sustains partial correction of WBCT 1.5- to 2-fold longer than rFVIII. rFVIIIFc was well tolerated in both species. Thus, the rescue of FVIII by Fc fusion to provide prolonged protection presents a novel pathway for FVIII catabolism, and warrants further investigation.


Sujet(s)
Facteur VIII/pharmacocinétique , Hémophilie A/métabolisme , Antigènes d'histocompatibilité de classe I/pharmacologie , Protéines de fusion recombinantes/pharmacocinétique , Animaux , Coagulants/pharmacocinétique , Coagulants/usage thérapeutique , Modèles animaux de maladie humaine , Maladies des chiens/traitement médicamenteux , Maladies des chiens/métabolisme , Chiens , Facteur VIII/composition chimique , Facteur VIII/génétique , Facteur VIII/usage thérapeutique , Cellules HEK293 , Période , Hémophilie A/traitement médicamenteux , Hémophilie A/anatomopathologie , Antigènes d'histocompatibilité de classe I/composition chimique , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/usage thérapeutique , Humains , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Récepteur Fc/composition chimique , Récepteur Fc/métabolisme , Récepteur Fc/usage thérapeutique , Protéines de fusion recombinantes/usage thérapeutique , Temps de coagulation
9.
Bioconjug Chem ; 23(3): 518-26, 2012 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-22263969

RÉSUMÉ

Atrial natriuretic peptide (ANP) may be a useful molecule for the treatment of cardiovascular diseases due to its potent natriuretic effects. In an effort to prolong the short in vivo half-life of ANP, fusions of the peptide to the Fc domain of IgG were generated using a semisynthetic methodology. Synthetic ANP peptides were synthesized with thioesters at either the N- or C-termini of the peptide and subsequently linked to the N-terminus of recombinantly expressed Fc using native chemical ligation. The linker length between the ANP and Fc moieties was varied among 2, 11, or 16 amino acids. In addition, either one ("monomeric") or two ("dimeric") ANP peptides were linked to Fc to study whether this modification had an effect on in vitro activity and/or in vivo half-life. The various constructs were studied for in vitro activity using a cell-based cGMP assay. The ANP-Fc fusion constructs were between 16- and ∼375-fold weaker than unconjugated ANP in this assay, and a trend was observed where the most potent conjugates were those with longer linkers and in the dimeric configuration. The pharmacokinetics of several constructs were assessed in rats, and the half-life of the ANP-Fc's were found to be approximately 2 orders of magnitude longer than that of the unconjugated peptide. There was no significant difference in terminal half-life between the monomeric and dimeric constructs (2.8-5.5 h), but a trend was observed where the C(max) of the monomeric constructs was approximately 3-fold higher than that of the dimeric constructs, although the origin of this effect is not understood. These novel ANP-Fc fusion constructs hold promise for future therapeutic application in the treatment of cardiovascular diseases.


Sujet(s)
Facteur atrial natriurétique/pharmacocinétique , Fragments Fc des immunoglobulines/composition chimique , Animaux , Facteur atrial natriurétique/composition chimique , Lignée cellulaire , Chromatographie sur gel , Test ELISA , Femelle , Période , Humains , Techniques in vitro , Rats , Rat Wistar , Protéines de fusion recombinantes/synthèse chimique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/pharmacocinétique
10.
J Interferon Cytokine Res ; 32(4): 178-84, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-22191463

RÉSUMÉ

Currently, products containing interferon beta (IFNß) are injected either intramuscularly or subcutaneously. To avoid the necessity of injection, we developed a novel monomeric Fc fusion protein of IFNß (IFNßFc) that is absorbed via an immunoglobulin transport system present in the upper and central airways upon administration of the drug as an inhaled aerosol. The systemic absorption of IFNßFc through the lung in non-human primates, at deposited doses of 1, 3, and 10 µg/kg, was compared to the absorption of a single 3 µg/kg dose of IFNß-1a (Avonex®) subcutaneously administered. IFNßFc was well absorbed through the lung, displaying dose proportional increases in serum concentrations, and was biologically active, as shown by increases in plasma neopterin levels. The circulating half-life of IFNßFc was ∼3 times longer (∼30 h) than that of IFNß-1a, (8-9 h). At approximately equimolar doses of IFNßFc (10 µg/kg) and IFNß-1a (3 µg/kg), the stimulation of neopterin over background levels was approximately equivalent, demonstrating that the longer half-life of IFNßFc compensated for the lower relative specific antiviral activity of IFNßFc measured in vitro. In conclusion, IFNßFc was efficiently absorbed after pulmonary delivery in non-human primates, retained its biological activity, and may offer a convenient alternative to injectable IFNß.


Sujet(s)
Fragments Fc des immunoglobulines/administration et posologie , Fragments Fc des immunoglobulines/pharmacologie , Immunoglobulines/métabolisme , Interféron bêta/administration et posologie , Interféron bêta/pharmacologie , Poumon/effets des médicaments et des substances chimiques , Poumon/immunologie , Protéines de fusion recombinantes/administration et posologie , Protéines de fusion recombinantes/pharmacologie , Administration par inhalation , Animaux , Chromatographie sur gel , Électrophorèse sur gel de polyacrylamide , Femelle , Humains , Fragments Fc des immunoglobulines/biosynthèse , Injections sous-cutanées , Interféron bêta-1a , Interféron bêta/biosynthèse , Interféron bêta/pharmacocinétique , Macaca fascicularis/sang , Mâle , Néoptérine/sang , Transport des protéines/effets des médicaments et des substances chimiques , Protéines de fusion recombinantes/biosynthèse , Protéines de fusion recombinantes/pharmacocinétique
11.
Bioorg Med Chem Lett ; 21(21): 6332-5, 2011 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-21920737

RÉSUMÉ

Peptides targeting the human neonatal Fc receptor (FcRn) were conjugated to poly(ethylene glycol) (PEG) polymers to study their effect on inhibition of the IgG:FcRn protein-protein interaction both in vitro and in mice. Both linear (5-40kDa) and branched (20, 40kDa) PEG aldehydes were conjugated to an amine-containing linker of a homodimeric anti-FcRn peptide using reductive alkylation chemistry. It was found that conjugation of PEG to the peptide compromised the in vitro activity, with larger and branched PEGs causing the most dramatic losses in activity. The conjugates were evaluated in transgenic mice for their ability to accelerate the catabolism of human IgG. Optimal pharmacodynamic properties were observed with PEG-peptide conjugates that contained 20-40kDa linear PEGs and a 20kDa branched PEG. The optimal PEG-peptide conjugates were more effective in vivo than the unconjugated peptide control on a mole:mole and mg/kg basis, and represent potential new longer-acting peptide therapeutics for the treatment of humorally-mediated autoimmune disease.


Sujet(s)
Peptides/pharmacologie , Polyéthylène glycols/composition chimique , Récepteur Fc/antagonistes et inhibiteurs , Animaux , Électrophorèse sur gel de polyacrylamide , Test ELISA , Humains , Immunoglobuline G/métabolisme , Souris , Souris transgéniques , Peptides/antagonistes et inhibiteurs , Récepteur Fc/métabolisme
12.
J Med Chem ; 53(4): 1587-96, 2010 Feb 25.
Article de Anglais | MEDLINE | ID: mdl-20092334

RÉSUMÉ

The neonatal Fc receptor, FcRn, regulates the half-life of IgG in vivo and may be a target in the treatment of autoimmune disease. Monomeric peptide antagonists of the human IgG-human FcRn interaction were dimerized using three different synthetic methodologies: thiol/alkyl halide coupling of unprotected peptides, reductive alkylation of unprotected peptides, and on-resin amide bond formation with protected peptides. It was found that dimerization of monomeric peptides increased the in vitro activity of the peptide monomers more than 200-fold. Human IgG catabolism experiments in human FcRn transgenic mice were used to assess the in vivo activity of peptide dimers that possessed different linkers, cyclizations, and affinities for FcRn. Overall, it was found that the linker joining two monomeric peptides had only a minor effect on the in vitro potency but that in vitro potency was predictive of in vivo activity.


Sujet(s)
Antigènes d'histocompatibilité de classe I/métabolisme , Immunoglobuline G/métabolisme , Oligopeptides/synthèse chimique , Peptides cycliques/synthèse chimique , Récepteur Fc/métabolisme , Animaux , Dimérisation , Antigènes d'histocompatibilité de classe I/génétique , Humains , Souris , Souris transgéniques , Oligopeptides/composition chimique , Oligopeptides/pharmacologie , Peptides cycliques/composition chimique , Peptides cycliques/pharmacologie , Récepteur Fc/génétique , Relation structure-activité
13.
Blood ; 115(10): 2057-64, 2010 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-20056791

RÉSUMÉ

Treatment of hemophilia B requires frequent infusions of factor IX (FIX) to prophylax against bleeding episodes. Hemophilia B management would benefit from a FIX protein with an extended half-life. A recombinant fusion protein (rFIXFc) containing a single FIX molecule attached to the Fc region of immunoglobulin G was administered intravenously and found to have an extended half-life, compared with recombinant FIX (rFIX) in normal mice, rats, monkeys, and FIX-deficient mice and dogs. Recombinant FIXFc protein concentration was determined in all species, and rFIXFc activity was measured in FIX-deficient animals. The half-life of rFIXFc was approximately 3- to 4-fold longer than that of rFIX in all species. In contrast, in mice in which the neonatal Fc receptor (FcRn) was deleted, the half-life of rFIXFc was similar to rFIX, confirming the increased circulatory time was due to protection of the rFIXFc via the Fc/FcRn interaction. Whole blood clotting time in FIX-deficient mice was corrected through 144 hours for rFIXFc, compared with 72 hours for rFIX; similar results were observed in FIX-deficient dogs. Taken together, these studies show the enhanced pharmacodynamic and pharmacokinetic properties of the rFIXFc fusion protein and provide the basis for evaluating rFIXFc in patients with hemophilia B.


Sujet(s)
Coagulation sanguine/effets des médicaments et des substances chimiques , Facteur IX/pharmacocinétique , Fragments Fc des immunoglobulines/pharmacologie , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/pharmacocinétique , Animaux , Temps de saignement , Coagulation sanguine/génétique , Cellules cultivées , Maladies des chiens/sang , Maladies des chiens/traitement médicamenteux , Chiens , Évaluation préclinique de médicament , Facteur IX/génétique , Facteur IX/métabolisme , Facteur IX/physiologie , Facteur IX/usage thérapeutique , Femelle , Hémophilie B/sang , Hémophilie B/traitement médicamenteux , Hémophilie B/médecine vétérinaire , Humains , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/métabolisme , Fragments Fc des immunoglobulines/usage thérapeutique , Macaca fascicularis , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Multimérisation de protéines , Rats , Protéines de fusion recombinantes/usage thérapeutique , Facteurs temps
14.
AAPS J ; 11(3): 432-4, 2009 Sep.
Article de Anglais | MEDLINE | ID: mdl-19499344

RÉSUMÉ

The neonatal Fc receptor, FcRn, is responsible for controlling the half-life of IgG antibodies. As a result, inhibitors of FcRn have been investigated as a possible way to modulate IgG half-lives. Such inhibitors could have possible applications in reducing autoantibody levels in autoimmune disease states. To date, monoclonal antibodies, engineered Fc domains, and short peptides have been reported to inhibit FcRn function and modulate IgG half-lives in vivo.


Sujet(s)
Immunoglobuline G/métabolisme , Récepteur Fc/antagonistes et inhibiteurs , Animaux , Anticorps/génétique , Anticorps/immunologie , Période , Antigènes d'histocompatibilité de classe I/métabolisme , Humains , Immunoglobuline G/immunologie , Liaison aux protéines , Ingénierie des protéines , Récepteur Fc/métabolisme
15.
Proc Natl Acad Sci U S A ; 105(7): 2337-42, 2008 Feb 19.
Article de Anglais | MEDLINE | ID: mdl-18272495

RÉSUMÉ

The neonatal Fc receptor FcRn provides IgG molecules with their characteristically long half-lives in vivo by protecting them from intracellular catabolism and then returning them to the extracellular space. Other investigators have demonstrated that mice lacking FcRn are protected from induction of various autoimmune diseases, presumably because of the accelerated catabolism of pathogenic IgGs in the animals. Therefore, targeting FcRn with a specific inhibitor may represent a unique approach for the treatment of autoimmune disease or other diseases where the reduction of pathogenic IgG will have a therapeutic benefit. Using phage display peptide libraries, we screened for ligands that bound to human FcRn (hFcRn) and discovered a consensus peptide sequence that binds to hFcRn and inhibits the binding of human IgG (hIgG) in vitro. Chemical optimization of the phage-identified sequences yielded the 26-amino acid peptide dimer SYN1436, which is capable of potent in vitro inhibition of the hIgG-hFcRn interaction. Administration of SYN1436 to mice transgenic for hFcRn induced an increase in the rate of catabolism of hIgG in a dose-dependent manner. Treatment of cynomolgus monkeys with SYN1436 led to a reduction of IgG by up to 80% without reducing serum albumin levels that also binds to FcRn. SYN1436 and related peptides thus represent a previously uncharacterized family of potential therapeutic agents for the treatment of humorally mediated autoimmune and other diseases.


Sujet(s)
Antigènes d'histocompatibilité de classe I/immunologie , Fragments Fc des immunoglobulines/immunologie , Immunoglobuline G/immunologie , Macaca fascicularis/immunologie , Peptides/immunologie , Récepteur Fc/antagonistes et inhibiteurs , Récepteur Fc/immunologie , Albumines/métabolisme , Séquence d'acides aminés , Animaux , Lignée cellulaire , Antigène HLA-A2/immunologie , Antigène HLA-A2/métabolisme , Humains , Immunoglobuline A/immunologie , Immunoglobuline M/immunologie , Macaca fascicularis/métabolisme , Données de séquences moléculaires , Peptides/composition chimique , Liaison aux protéines , Solubilité , Résonance plasmonique de surface
16.
BioDrugs ; 20(3): 151-60, 2006.
Article de Anglais | MEDLINE | ID: mdl-16724863

RÉSUMÉ

The delivery of therapeutic proteins by noninvasive routes of administration has been a challenging goal, hence current modes of delivery generally require injections. However, we have recently shown that a naturally occurring receptor, the neonatal Fc receptor (FcRn) can be utilized to carry aerosolized therapeutic proteins conjugated to a portion of its respective ligand (Fc domain of immunoglobulin G) across epithelial cells of the lung to effectively deliver biologically active molecules to the bloodstream. First-generation dimeric Fc fusion molecules were successfully transported by the pulmonary route and biologic activity was demonstrated in both non-human primates and human volunteers. Continuing efforts to improve transport efficiency have led to the development of an alternate configuration of Fc fusion proteins with improved characteristics. These second generation Fc fusion molecules are monomeric with respect to the therapeutic protein and dimeric with respect to the Fc region, and have been termed Fc fusion 'monomers'. Several different Fc fusion monomers have demonstrated improved transport efficiency, achieving high bioavailabilities for pulmonary delivery in non-human primates. While the traditional dimeric Fc fusion molecule generally increases the half-life compared with the unconjugated effector molecule, the monomer configuration has been shown to result in an even greater extension of the circulating half-life, which improves pharmacokinetic parameters for protein therapeutics, whether administered by pulmonary delivery or injection. Finally, many of the Fc monomer fusions have enhanced biologic activity compared with the dimeric configuration. Because of these many advantages, the monomer configuration promises to be an enabling advance to achieve clinically relevant, noninvasive delivery with potentially less frequent administration regimens for a broad range of protein therapeutics. In addition, molecules that are comprised of heterodimeric subunits or multi-subunit complexes can also be constructed as Fc fusions that result in a molecule with enhanced pharmacokinetics and greater bioactivity. Several examples of novel Fc fusion proteins, both monomer and heterodimer are described herein.


Sujet(s)
Érythropoïétine/métabolisme , Facteur IX/métabolisme , Antigènes d'histocompatibilité de classe I/métabolisme , Fragments Fc des immunoglobulines/métabolisme , Interféron alpha/métabolisme , Interféron bêta/métabolisme , Poumon/métabolisme , Récepteur Fc/métabolisme , Protéines de fusion recombinantes/pharmacocinétique , Animaux , Systèmes de délivrance de médicaments , Humains , Liaison aux protéines , Transport des protéines
17.
Proc Natl Acad Sci U S A ; 101(26): 9763-8, 2004 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-15210944

RÉSUMÉ

Administration of therapeutic proteins by methods other than injection is limited, in part, by inefficient penetration of epithelial barriers. Therefore, unique approaches to breaching these barriers are needed. The neonatal constant region fragment (Fc) receptor (FcRn), which is responsible for IgG transport across the intestinal epithelium in newborn rodents, is expressed in epithelial cells in adult humans and non-human primates. Here we show that FcRn-mediated transport is functional in the lung of non-human primates and that this transport system can be used to deliver erythropoietin (Epo) when it is conjugated to the Fc domain of IgG1. FcRn-dependent absorption was more efficient when the EpoFc fusion protein was deposited predominantly in the upper and central airways of the lung, where epithelial expression of FcRn was most prominently detected. To optimize fusion protein absorption in the lung, we created a recombinant "monomeric-Epo" Fc fusion protein comprised of a single molecule of Epo conjugated to a dimeric Fc. This fusion protein exhibited enhanced pharmacokinetic and pharmacodynamic properties. The bioavailability of the EpoFc monomer when delivered through the lung was approximately equal to that reported for unconjugated Epo delivered s.c. in humans. These studies show that FcRn can be harnessed to noninvasively deliver bioactive proteins into the systemic circulation in therapeutic quantities.


Sujet(s)
Érythropoïétine/métabolisme , Érythropoïétine/pharmacocinétique , Fragments Fc des immunoglobulines/métabolisme , Poumon/métabolisme , Macaca fascicularis/métabolisme , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/pharmacocinétique , Absorption , Animaux , Dimérisation , Systèmes de délivrance de médicaments , Érythropoïétine/composition chimique , Érythropoïétine/génétique , Humains , Fragments Fc des immunoglobulines/composition chimique , Fragments Fc des immunoglobulines/génétique , Immunoglobuline G/composition chimique , Immunoglobuline G/génétique , Immunoglobuline G/métabolisme , Poumon/cytologie , Liaison aux protéines , Transport des protéines , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Respiration , Solubilité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE