Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Clin Med ; 11(3)2022 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-35160081

RÉSUMÉ

People living with sickle cell disease (SCD) face intermittent acute pain episodes due to vaso-occlusion primarily treated palliatively with opioids. Hemolysis of sickle erythrocytes promotes release of heme, which activates inflammatory cell adhesion proteins on endothelial cells and circulating cells, promoting vaso-occlusion. In this study, plasma-derived hemopexin inhibited heme-mediated cellular externalization of P-selectin and von Willebrand factor, and expression of IL-8, VCAM-1, and heme oxygenase-1 in cultured endothelial cells in a dose-responsive manner. In the Townes SCD mouse model, intravenous injection of free hemoglobin induced vascular stasis (vaso-occlusion) in nearly 40% of subcutaneous blood vessels visualized in a dorsal skin-fold chamber. Hemopexin administered intravenously prevented or relieved stasis in a dose-dependent manner. Hemopexin showed parallel activity in relieving vascular stasis induced by hypoxia-reoxygenation. Repeated IV administration of hemopexin was well tolerated in rats and non-human primates with no adverse findings that could be attributed to human hemopexin. Hemopexin had a half-life in wild-type mice, rats, and non-human primates of 80-102 h, whereas a reduced half-life of hemopexin in Townes SCD mice was observed due to ongoing hemolysis. These data have led to a Phase 1 clinical trial of hemopexin in adults with SCD, which is currently ongoing.

2.
Blood Coagul Fibrinolysis ; 33(1): 56-60, 2022 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-34267062

RÉSUMÉ

Insufficiency of ADAMTS13 (a disintegrin and metalloprotease with thrombospondin motif repeats-13) is the cause of thrombotic thrombocytopenic purpura (TTP) and contributes in microangiopathy in sickle cell disease (SCD). Recombinant ADAMTS13 effectively cleaves prothrombotic ultra-large von Willebrand factor (VWF) multimers. It is being tested as replacement therapy for TTP, and at supra-physiologic concentrations, for moderating vaso-occlusive crisis in SCD. Deficiencies of VWF, or concomitant treatment with antithrombotic drugs, could pose risks for increased bleeds in these patient populations. The purpose of the experiments was to evaluate the potential of exaggerated pharmacology and temporary bleeding risks associated with rADAMTS13 administration. We utilized safety studies in monkey and tested the effects of administering maximum-feasible doses of rADAMTS13 on nonclinical safety and spontaneous or aggressive bleeds in the rat model. Evaluation of pharmacokinetics, toxicity profiles, and challenge in a tail-tip bleeding model show that treatment with rADAMTS13 did not increase bleeding tendency, either alone, or in combination with enoxaparin or acetylsalicylic-acid. These novel findings demonstrate absence of rADAMTS13 exaggerated pharmacology without spontaneous or aggravated bleeds even at supra-physiologic (>100-fold) plasma concentrations.


Sujet(s)
Protéines ADAM , Purpura thrombotique thrombocytopénique , Protéine ADAMTS13 , Animaux , Haplorhini , Humains , Rats , Facteur de von Willebrand
3.
J Pharmacol Exp Ther ; 371(1): 95-105, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31366602

RÉSUMÉ

Extended half-life (EHL) factor therapies are needed to reduce the burden of prophylaxis and improve treatment adherence in patients with hemophilia. BAX 826 is a novel polysialylated full-length recombinant factor VIII [polysialyic acid (PSA) rFVIII] with improved pharmacokinetics (PK), prolonged pharmacology, and maintained safety attributes to enable longer-acting rFVIII therapy. In factor VIII (FVIII)-deficient hemophilic mice, PSArFVIII showed a substantially higher mean residence time (>2-fold) and exposure (>3-fold), and prolonged efficacy in tail-bleeding experiments (48 vs. 30 hours) compared with unmodified recombinant FVIII (rFVIII), as well as a potentially favorable immunogenicity profile. Reduced binding to a scavenger receptor (low-density lipoprotein receptor-related protein 1) and von Willebrand factor (VWF) as well as a largely VWF-independent circulation time in mice provide a rationale for prolonged BAX 826 activity. The significantly improved PK profile versus rFVIII was confirmed in cynomolgus monkeys [mean residence time: 23.4 vs. 10.1 hours; exposure (area under the curve from time 0 to infinity): 206 vs. 48.2 IU/ml⋅h] and is in line with results from rodent studies. Finally, safety and toxicity evaluations did not indicate increased thrombogenic potential, and repeated administration of BAX 826 to monkeys and rats was well tolerated. The favorable profile and mechanism of this novel experimental therapeutic demonstrated all of the requirements for an EHL-rFVIII candidate, and thus BAX 826 was entered into clinical assessment for the treatment of hemophilia A. SIGNIFICANCE STATEMENT: Prolongation of FVIII half-life aims to reduce the burden of prophylaxis and improve treatment outcomes in patients with hemophilia. This study shows that polysialylation of PSArFVIII resulted in prolongations of rFVIII circulation time and procoagulant activity, together with a favorable nonclinical safety profile of the experimental therapeutic.


Sujet(s)
Facteur VIII/usage thérapeutique , Hémophilie A/traitement médicamenteux , Absorption physiologique , Animaux , Essais cliniques comme sujet , Évaluation préclinique de médicament , Facteur VIII/effets indésirables , Facteur VIII/pharmacocinétique , Femelle , Période , Humains , Macaca fascicularis , Mâle , Acide N-acétyl-neuraminique/composition chimique , Liaison aux protéines , Rats , Récepteurs éboueurs/métabolisme , Facteur de von Willebrand/métabolisme
4.
PLoS One ; 6(4): e18577, 2011 Apr 07.
Article de Anglais | MEDLINE | ID: mdl-21490925

RÉSUMÉ

BACKGROUND: H5N1 influenza vaccines, including live intranasal, appear to be relatively less immunogenic compared to seasonal analogs. The main influenza virus surface glycoprotein hemagglutinin (HA) of highly pathogenic avian influenza viruses (HPAIV) was shown to be more susceptible to acidic pH treatment than that of human or low pathogenic avian influenza viruses. The acidification machinery of the human nasal passageway in response to different irritation factors starts to release protons acidifying the mucosal surface (down to pH of 5.2). We hypothesized that the sensitivity of H5 HA to the acidic environment might be the reason for the low infectivity and immunogenicity of intranasal H5N1 vaccines for mammals. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate that original human influenza viruses infect primary human nasal epithelial cells at acidic pH (down to 5.4), whereas H5N1 HPAIVs lose infectivity at pH ≤ 5.6. The HA of A/Vietnam/1203/04 was modified by introducing the single substitution HA2 58K→I, decreasing the pH of the HA conformational change. The H5N1 reassortants containing the indicated mutation displayed an increased resistance to acidic pH and high temperature treatment compared to those lacking modification. The mutation ensured a higher viral uptake as shown by immunohistochemistry in the respiratory tract of mice and 25 times lower mouse infectious dose50. Moreover, the reassortants keeping 58K→I mutation designed as a live attenuated vaccine candidate lacking an NS1 gene induced superior systemic and local antibody response after the intranasal immunization of mice. CONCLUSION/SIGNIFICANCE: Our finding suggests that an efficient intranasal vaccination with a live attenuated H5N1 virus may require a certain level of pH and temperature stability of HA in order to achieve an optimal virus uptake by the nasal epithelial cells and induce a sufficient immune response. The pH of the activation of the H5 HA protein may play a substantial role in the infectivity of HPAIVs for mammals.


Sujet(s)
Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H5N1 du virus de la grippe A/immunologie , Vaccins antigrippaux/génétique , Vaccins antigrippaux/immunologie , Infections à Orthomyxoviridae/génétique , Infections à Orthomyxoviridae/immunologie , Administration par voie nasale , Animaux , Chlorocebus aethiops , Test ELISA , Femelle , Humains , Immunoglobuline A/sang , Immunoglobuline G/sang , Sous-type H5N1 du virus de la grippe A/pathogénicité , Vaccins antigrippaux/administration et posologie , Souris , Mutation , Vaccins atténués/administration et posologie , Vaccins atténués/génétique , Vaccins atténués/immunologie , Cellules Vero
5.
Vaccine ; 29(19): 3517-24, 2011 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-21406268

RÉSUMÉ

The isolation and cultivation of human influenza viruses in embryonated hen eggs or cell lines often leads to amino acid substitutions in the haemagglutinin (HA) molecule. We found that the propagation of influenza A H3N2 viruses on Vero cells may trigger the appearance of HA destabilising mutations, affecting viral resistance to low pH or high temperature treatment. Two ΔNS1 reassortants, containing the HA sequences identical to the original human H3N2 influenza virus isolates were constructed. Passages of these viruses on Vero cells led to the appearance of single mutations in the HA(1) L194P or HA(2) G75R subunits that impaired virus stability. The original HA sequences and the stable phenotypes of the primary isolates were preserved if reassortants were passaged by infection at pH 5.6 and cultivation in medium at pH 6.5. Corresponding ΔNS1 reassortants were compared for their immunogenicity in ferrets upon intranasal immunisation. Vaccine candidates containing HA mutations demonstrated significantly lower immunogenicity compared to those without mutations. Thus, the retaining of the original HA sequences of human viruses during vaccine production might be crucial for the efficacy of live attenuated influenza vaccines.


Sujet(s)
Glycoprotéine hémagglutinine du virus influenza/génétique , Sous-type H3N2 du virus de la grippe A/génétique , Administration par voie nasale , Animaux , Anticorps neutralisants/sang , Anticorps antiviraux/sang , Lignée cellulaire , Chlorocebus aethiops , Femelle , Furets , Glycoprotéine hémagglutinine du virus influenza/immunologie , Concentration en ions d'hydrogène , Immunité humorale , Immunisation , Sous-type H3N2 du virus de la grippe A/immunologie , Vaccins antigrippaux/génétique , Vaccins antigrippaux/immunologie , Mâle , Mutation , Virus recombinants/génétique , Virus recombinants/immunologie , Cellules Vero
6.
Cancer Biol Ther ; 10(6): 592-9, 2010 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-20647775

RÉSUMÉ

Previously we have developed a prototype for conditionally replicating oncolytic influenza A virus which is based on deletions in the non-structural (NS1) protein. Multi-cycle replication of influenza A virus in malignant tissue is critically dependent on a protease which cleaves the viral entry protein. Here we demonstrate that the malignant colon cancer cell lines Caco-2, HT-29 and SW-620 can endogenously provide a virus-activating protease, which allows lytic multi-cycle replication of NS1 deletion viruses in those cancer cells in vitro. The oncolytic potency of an influenza NS1 deletion virus (NS1-80) was further tested in SCID mice bearing HT-29 derived tumors. The intra-tumoral injection of live, but not of heat inactivated NS1-80 virus significantly inhibited progression of established tumors. We conclude that a selected set of human cancer expressing virus activating- proteases will be a preferred target for oncolytic tumor therapy using influenza A virus mutants.


Sujet(s)
Tumeurs du côlon/enzymologie , Virus de la grippe A/métabolisme , Virus oncolytiques/métabolisme , Peptide hydrolases/métabolisme , Protéines virales non structurales/métabolisme , Animaux , Cellules Caco-2 , Lignée cellulaire tumorale , Chlorocebus aethiops , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/virologie , Femelle , Cellules HCT116 , Cellules HT29 , Interactions hôte-pathogène , Humains , Virus de la grippe A/génétique , Virus de la grippe A/physiologie , Souris , Souris SCID , Mutation , Tumeurs expérimentales/enzymologie , Tumeurs expérimentales/anatomopathologie , Tumeurs expérimentales/virologie , Virus oncolytiques/génétique , Virus oncolytiques/physiologie , Infections à Orthomyxoviridae/enzymologie , Infections à Orthomyxoviridae/anatomopathologie , Infections à Orthomyxoviridae/virologie , Peptide hydrolases/génétique , Transplantation hétérologue , Cellules Vero , Protéines virales non structurales/génétique
7.
PLoS One ; 4(6): e5984, 2009 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-19543385

RÉSUMÉ

BACKGROUND: We developed a novel intranasal influenza vaccine approach that is based on the construction of replication-deficient vaccine viruses that lack the entire NS1 gene (DeltaNS1 virus). We previously showed that these viruses undergo abortive replication in the respiratory tract of animals. The local release of type I interferons and other cytokines and chemokines in the upper respiratory tract may have a "self-adjuvant effect", in turn increasing vaccine immunogenicity. As a result, DeltaNS1 viruses elicit strong B- and T- cell mediated immune responses. METHODOLOGY/PRINCIPAL FINDINGS: We applied this technology to the development of a pandemic H5N1 vaccine candidate. The vaccine virus was constructed by reverse genetics in Vero cells, as a 5:3 reassortant, encoding four proteins HA, NA, M1, and M2 of the A/Vietnam/1203/04 virus while the remaining genes were derived from IVR-116. The HA cleavage site was modified in a trypsin dependent manner, serving as the second attenuation factor in addition to the deleted NS1 gene. The vaccine candidate was able to grow in the Vero cells that were cultivated in a serum free medium to titers exceeding 8 log(10) TCID(50)/ml. The vaccine virus was replication deficient in interferon competent cells and did not lead to viral shedding in the vaccinated animals. The studies performed in three animal models confirmed the safety and immunogenicity of the vaccine. Intranasal immunization protected ferrets and mice from being infected with influenza H5 viruses of different clades. In a primate model (Macaca mulatta), one dose of vaccine delivered intranasally was sufficient for the induction of antibodies against homologous A/Vietnam/1203/04 and heterologous A/Indonesia/5/05 H5N1 strains. CONCLUSION/SIGNIFICANCE: Our findings show that intranasal immunization with the replication deficient H5N1 DeltaNS1 vaccine candidate is sufficient to induce a protective immune response against H5N1 viruses. This approach might be attractive as an alternative to conventional influenza vaccines. Clinical evaluation of DeltaNS1 pandemic and seasonal influenza vaccine candidates are currently in progress.


Sujet(s)
Sous-type H5N1 du virus de la grippe A/génétique , Vaccins antigrippaux/usage thérapeutique , Administration par voie nasale , Animaux , Bronches/cytologie , Lignée cellulaire , Poulets , Chlorocebus aethiops , Chiens , Évaluation préclinique de médicament , Cellules épithéliales/cytologie , Furets , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Humains , Vaccins antigrippaux/métabolisme , Macrophages/métabolisme , Macrophages/virologie , Souris , Cellules Vero , Réplication virale
8.
Vaccine ; 27(21): 2851-7, 2009 May 11.
Article de Anglais | MEDLINE | ID: mdl-19366569

RÉSUMÉ

We discovered a unique, single amino acid mutation in the influenza B M1 protein promoting viral growth of NS1 truncation mutants in Vero cells. Due to this mutation, we were able to generate an influenza B virus lacking the complete NS1 open reading frame (DeltaNS1-B virus) by reverse genetics, which was growing to titers of 8log(10)TCID(50)/ml in a Vero cell culture-based micro-carrier fermenter. The DeltaNS1-B vaccine candidate was attenuated in IFN-competent hosts such as human alveolar epithelial cells (A549) similar to influenza A DeltaNS1 viruses. In ferrets, the DeltaNS1-B virus was replication-deficient and did not provoke any clinical symptoms. Importantly, a single intranasal immunization of ferrets at a dose as low as 6 log(10)TCID(50)/animal induced a significant HAI response and provided protection against challenge with wild-type influenza B virus. So far, the lack of a DeltaNS1-B virus component growing to high titers in cell culture has been limiting the possibility to formulate a trivalent vaccine based on deletion of the NS1 gene. Our study closes this gap and paves the way for the clinical evaluation of a seasonal, trivalent, live replication-deficient DeltaNS1 intranasal influenza vaccine.


Sujet(s)
Virus influenza B/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/immunologie , Protéines virales non structurales/immunologie , Administration par voie nasale , Animaux , Séquence nucléotidique , Lignée cellulaire , Chlorocebus aethiops , Furets/immunologie , Humains , Virus influenza B/génétique , Virus influenza B/métabolisme , Mutation/génétique , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Vaccins atténués/génétique , Vaccins atténués/immunologie , Vaccins atténués/métabolisme , Protéines virales non structurales/génétique , Protéines virales non structurales/métabolisme , Réplication virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE