Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 202(5): 1582-1594, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30683704

RESUMEN

The long serum t 1/2 of IgGs is ensured by their interaction with the neonatal Fc receptor (FcRn), which salvages IgG from intracellular degradation. Fc glycosylation is thought not to influence FcRn binding and IgG longevity in vivo. In this article, we demonstrate that hypersialylation of asparagine 297 (N297) enhances IgG serum persistence. This polarized glycosylation is achieved using a novel Fc mutation, a glutamate residue deletion at position 294 (Del) that endows IgGs with an up to 9-fold increase in serum lifespan. The strongest impact was observed when the Del was combined with Fc mutations improving FcRn binding (Del-FcRn+). Enzymatic desialylation of a Del-FcRn+ mutant or its production in a cell line unable to hypersialylate reduced the in vivo serum t 1/2 of the desialylated mutants to that of native FcRn+ mutants. Consequently, our study proves that sialylation of the N297 sugar moiety has a direct impact on human IgG serum persistence.


Asunto(s)
Anticuerpos/sangre , Anticuerpos/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/sangre , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/sangre , Inmunoglobulina G/uso terapéutico , Animales , Anticuerpos/química , Células HEK293 , Semivida , Humanos , Inmunoglobulina G/química , Ratones , Ratones Noqueados
2.
Haematologica ; 105(9): 2335-2340, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054058

RESUMEN

Rendering coagulation factor X sensitive to thrombin was proposed as a strategy that can bypass the need for factor VIII. In this paper, this non-replacement strategy was evaluated in vitro and in vivo in its ability to correct factor VIII but also factor IX, X and XI deficiencies. A novel modified factor X, named Actiten, was generated and produced in the HEK293F cell line. The molecule possesses the required post-translational modifications, partially keeps its ability to be activated by RVV-X, factor VIIa/tissue factor, factor VIIIa/factor IXa and acquires the ability to be activated by thrombin. The potency of the molecule was evaluated in respective deficient plasmas or hemophilia A plasmas, for some with inhibitors. Actiten corrects dose dependently all the assayed deficient plasmas. It is able to normalize the thrombin generation at 20 µg/mL showing however an increased lagtime. It was then assayed in a rabbit antibody-induced model of hemophilia A where, in contrast to recombinant factor X wild-type, it normalized the bleeding time and the loss of hemoglobin. No sign of thrombogenicity was observed and the generation of activated factor X was controlled by the anticoagulation pathway in all performed coagulation assays. This data indicates that Actiten may be considered as a possible non replacement factor to treat hemophilia's with the advantage of being a zymogen correcting bleedings only when needed.


Asunto(s)
Factor X , Hemofilia A , Animales , Factor IX , Factor VIII/genética , Factor VIIa , Factor X/genética , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Conejos , Trombina
3.
Br J Haematol ; 180(5): 715-720, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29363751

RESUMEN

Heparin anticoagulation followed by protamine reversal is commonly used in cardiopulmonary bypass (CPB). As an alternative to protamine, a recombinant inactive antithrombin (riAT) was designed as an antidote to heparin and was previously shown to be as potent as protamine in-vitro. In the present study, riAT was assessed for its ability to neutralize heparin after CPB in a rat model. After 60 min of CPB under heparin, rats received 5 mg/kg protamine, 37.5 mg/kg riAT or phosphate buffered saline (PBS) as placebo. Residual anticoagulant activity was assessed using the activated partial thromboplastin time assay before, and 10-30 min after reversion. Haemodynamic monitoring was performed and plasma histamine concentration was also measured. In this model, riAT appeared to be as efficient as protamine in neutralizing heparin. Ten minutes after injection, riAT and protamine both decreased heparin activity, to 1.8 ± 1.3 and 4.5 ± 1.4 u/ml, respectively (23.1 ± 5.1 u/ml in placebo group). Furthermore, evolution of mean carotid arterial pressure, heart rate and plasma histamine levels was comparable in rats treated with PBS or riAT, while protamine exhibited haemodynamic side effects and increased histamine plasma concentration. Thus, riAT could represent an advantage over protamine in CPB because it efficiently reverses heparin activity without negative effects on haemodynamic parameters and plasma histamine level.


Asunto(s)
Anticoagulantes/farmacología , Puente Cardiopulmonar , Antagonistas de Heparina/farmacología , Heparina/farmacología , Protaminas/farmacología , Animales , Antitrombinas/farmacología , Hemodinámica/efectos de los fármacos , Histamina/metabolismo , Masculino , Ratas Wistar
4.
Anal Biochem ; 491: 52-4, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26302360

RESUMEN

Rapid and efficient structural analysis is key to the development of new monoclonal antibodies. We have developed a fast and easy process to obtain mass spectrometry profiles of antibodies from culture supernatant. Treatment of the supernatant with IdeS generates three fragments of 25 kDa that can be analyzed by liquid chromatography-mass spectrometry time-of-flight (LC-MS TOF) in one run: LC, Fd, and Fc/2. This process gives rapid access to isoform and glycoform profiles. To specifically measure the fucosylation yield, we included a one-pot treatment with EndoS that removes the distal glycan heterogeneity. Our process was successfully compared with high-performance capillary electrophoresis with laser-induced fluorescence detection (HPCE-LIF), currently considered as the "gold standard" method.


Asunto(s)
Anticuerpos Monoclonales/análisis , Cromatografía Líquida de Alta Presión , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Electroforesis Capilar , Glicosilación , Isoformas de Proteínas/análisis , Espectrometría de Fluorescencia
5.
BMC Biotechnol ; 11: 1, 2011 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-21208406

RESUMEN

BACKGROUND: The rat hybridoma cell line YB2/0 appears a good candidate for the large-scale production of low fucose recombinant mAbs due to its lower expression of fut8 gene than other commonly used rodent cell lines. However, important variations of the fucose content of recombinant mAbs are observed in production culture conditions. To improve our knowledge on the YB2/0 fucosylation capacity, we have cloned and characterized the rat fut8 gene. RESULTS: The cDNAs encoding the rat α1,6-fucosyltransferase (FucT VIII) were cloned from YB2/0 cells by polymerase chain reaction-based and 5' RNA-Ligase-Mediated RACE methods. The cDNAs contain an open reading frame of 1728 bp encoding a 575 amino acid sequence showing 94% and 88% identity to human and pig orthologs, respectively. The recombinant protein expressed in COS-7 cells exhibits a α1,6-fucosyltransferase activity toward human asialo-agalacto-apotransferrin. The rat fut8 gene is located on chromosome 6 q and spans over 140 kbp. It contains 9 coding exons and four 5'-untranslated exons. FISH analysis shows a heterogeneous copy number of fut8 in YB2/0 nuclei with 2.8 ± 1.4 mean copy number. The YB2/0 fut8 gene is expressed as two main transcripts that differ in the first untranslated exon by the usage of distinct promoters and alternative splicing. Luciferase assays allow defining the minimal promoting regions governing the initiation of the two transcripts, which are differentially expressed in YB2/0 as shown by duplex Taqman QPCR analysis. Bioinformatics analysis of the minimal promoter regions upstream exons E-2 and E-3, governing the transcription of T1 and T2 transcripts, respectively, evidenced several consensus sequences for potential transcriptional repressors. Transient transfections of Rat2 cells with transcription factor expression vectors allowed identifying KLF15 as a putative repressor of T1 transcript in Rat2 cells. CONCLUSION: Altogether, these data contribute to a better knowledge of fut8 expression in YB2/0 that will be useful to better control the fucosylation of recombinant mAbs produced in these cells.


Asunto(s)
Fucosiltransferasas/genética , Animales , Secuencia de Bases , Células COS , Línea Celular , Núcleo Celular , Chlorocebus aethiops , Mapeo Cromosómico , Clonación Molecular , Biología Computacional , ADN Complementario/química , ADN Complementario/genética , ADN Complementario/metabolismo , Fucosiltransferasas/biosíntesis , Fucosiltransferasas/química , Dosificación de Gen , Hibridomas , Hibridación Fluorescente in Situ , Interfase , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Ratas , Estadísticas no Paramétricas
6.
J Am Heart Assoc ; 10(19): e016287, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34569248

RESUMEN

Background Atherosclerosis is a complex pathology in which dysfunctional endothelium, activated leucocytes, macrophages, and lipid-laden foam cells are implicated, and in which plaque disruption is driven by many putative actors. This study aimed to identify accurate targetable biomarkers using new in vivo approaches to propose tools for improved diagnosis and treatment. Methods and Results Human scFv (single-chain fragment variable) selected by in vivo phage display in a rabbit model of atherosclerosis was reformatted as scFv fused to the scFv-Fc (single-chain fragment variable fused to the crystallizable fragment of immunoglobulin G format) antibodies. Their reactivity was tested using flow cytometry and immunoassays, and aorta sections from animal models and human carotid and coronary artery specimens. A pool of atherosclerotic proteins from human endarterectomies was co-immunoprecipitated with the selected scFv-Fc followed by mass spectrometry for target identification. Near-infrared fluorescence imaging was performed in Apoe-/- mice after injection of an Alexa Fluor 647-labeled scFv-Fc-2c antibody produced in a baculovirus system with 2 additional cysteine residues (ie, 2c) for future coupling to nano-objects for theranostic applications. One scFv-Fc clone (P3) displayed the highest cross-reactivity against atherosclerotic lesion sections (rabbit, mouse, and human) and was chosen for translational development. Mass spectrometry identified galectin-3, a ß-galactoside-binding lectin, as the leader target. ELISA and immunofluorescence assays with a commercial anti-galectin-3 antibody confirmed this specificity. P3 scFv-Fc-2c specifically targeted atherosclerotic plaques in the Apoe-/- mouse model. Conclusions These results provide evidence that the P3 antibody holds great promise for molecular imaging of atherosclerosis and other inflammatory pathologies involving macrophages. Recently, galectin-3 was proposed as a high-value biomarker for the assessment of coronary and carotid atherosclerosis.


Asunto(s)
Aterosclerosis , Bacteriófagos , Placa Aterosclerótica , Anticuerpos de Cadena Única , Animales , Apolipoproteínas E , Aterosclerosis/diagnóstico , Aterosclerosis/genética , Biomarcadores , Galectina 3/genética , Humanos , Ratones , Conejos , Anticuerpos de Cadena Única/genética
7.
Front Immunol ; 12: 728322, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512662

RESUMEN

Novel molecules that directly target the neonatal Fc receptor (FcRn) and/or Fc gamma receptors (FcγRs) are emerging as promising treatments for immunoglobulin G (IgG)-dependent autoimmune pathologies. Mutated Fc regions and monoclonal antibodies that target FcRn are currently in clinical development and hold promise for reducing the levels of circulating IgG. Additionally, engineered structures containing multimeric Fc regions allow the dual targeting of FcRn and FcγRs; however, their tolerance needs to first be validated in phase I clinical studies. Here, for the first time, we have developed a modified monomeric recombinant Fc optimized for binding to all FcRns and FcγRs without the drawback of possible tolerance associated with FcγR cross-linking. A rational approach using Fc engineering allowed the selection of LFBD192, an Fc with a combination of six mutations that exhibits improved binding to human FcRn and FcγR as well as mouse FcRn and FcγRIV. The potency of LFBD192 was compared with that of intravenous immunoglobulin (IVIg), an FcRn blocker (Fc-MST-HN), and a trimeric Fc that blocks FcRn and/or immune complex-mediated cell activation through FcγR without triggering an immune reaction in several in vitro tests and validated in three mouse models of autoimmune disease.


Asunto(s)
Antirreumáticos/farmacología , Artritis Experimental/prevención & control , Autoinmunidad/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/farmacología , Receptores Fc/antagonistas & inhibidores , Receptores de IgG/antagonistas & inhibidores , Animales , Antirreumáticos/metabolismo , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Unión Competitiva , Complemento C5a/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Interleucina-2/metabolismo , Células Jurkat , Cinética , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fagocitosis/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Unión Proteica , Ingeniería de Proteínas , Receptores Fc/genética , Receptores Fc/inmunología , Receptores Fc/metabolismo , Receptores de IgG/genética , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Vías Secretoras , Transducción de Señal , Células THP-1
8.
Thromb Haemost ; 100(4): 670-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18841291

RESUMEN

The Fab-fragment of 6B4, a murine monoclonal antibody targeting the human platelet glycoprotein (GP) Ibalpha and blocking the binding of von Willebrand factor (VWF), is a powerful antithrombotic. In baboons, this was without side effects such as bleeding or thrombocytopenia. Recently, we developed a fully recombinant and humanized version of 6B4-Fab-fragment, h6B4-Fab, which maintains its inhibitory capacities in vitro and ex vivo after injection in baboons. We here investigated the antithrombotic properties, the effect on bleeding time and blood loss and initial pharmacokinetics of h6B4-Fab in baboons. The antithrombotic effect of h6B4-Fab on acute platelet-mediated thrombosis was studied in baboons where thrombus formation is induced at an injured and stenosed site of the femoral artery, allowing for cyclic flow reductions (CFRs) which are measured on an extracorporeal femoral arteriovenous shunt. Injection of 0.5 mg/kg h6B4-Fab significantly reduced the CFRs by 80%, whereas two extra injections, resulting in cumulative doses of 1.5 and 2.5 mg/kg, completely inhibited the CFRs. Platelet receptor occupancy, plasma concentrations and effects ex vivo were consistent with what was previously observed. Finally, minimal effects on bleeding time and blood loss, no spontaneous bleeding and no thrombocytopenia were observed. We therefore conclude that h6B4-Fab maintains the antithrombotic capacities of the murine 6B4-Fab, without causing side effects and therefore can be used for further development.


Asunto(s)
Plaquetas/inmunología , Arteria Femoral/lesiones , Complejo GPIb-IX de Glicoproteína Plaquetaria/inmunología , Trombosis/tratamiento farmacológico , Trombosis/inmunología , Enfermedad Aguda , Animales , Antibacterianos/farmacología , Tiempo de Sangría , Constricción Patológica , Modelos Animales de Enfermedad , Arteria Femoral/patología , Hemorragia/inducido químicamente , Hemorragia/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/efectos adversos , Fragmentos Fab de Inmunoglobulinas/inmunología , Ratones , Papio , Adhesividad Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/inmunología , Recuento de Plaquetas , Complejo GPIb-IX de Glicoproteína Plaquetaria/efectos adversos , Flujo Sanguíneo Regional , Ristocetina/farmacología , Estrés Mecánico , Trombocitopenia/inducido químicamente , Trombocitopenia/inmunología , Trombosis/fisiopatología
9.
Sci Rep ; 8(1): 15016, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30302027

RESUMEN

Atherosclerosis is a chronic, progressive inflammatory disease that may develop into vulnerable lesions leading to thrombosis. This pathology is characterized by the deposition of lipids within the arterial wall and infiltration of immune cells leading to amplification of inflammation. Nowadays there is a rising interest to assess directly the molecular and cellular components that underlie the clinical condition of stroke and myocardial infarction. Single chain fragment variable (scFv)-phages issuing from a human combinatorial library were selected on the lesions induced in a rabbit model of atherosclerosis after three rounds of in vivo phage display. We further implemented a high-throughput flow cytometry method on rabbit protein extracts to individually test one thousand of scFv-phages. Two hundred and nine clones were retrieved on the basis of their specificity for atherosclerotic proteins. Immunohistochemistry assays confirmed the robustness of the designed cytometry protocol. Sequencing of candidates demonstrated their high diversity in VH and VL germline usage. The large number of candidates and their diversity open the way in the discovery of new biomarkers. Here, we successfully showed the capacity of combining in vivo phage display and high-throughput cytometry strategies to give new insights in in vivo targetable up-regulated biomarkers in atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Técnicas de Visualización de Superficie Celular , Citometría de Flujo , Anticuerpos de Cadena Única/aislamiento & purificación , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica/métodos , Conejos , Anticuerpos de Cadena Única/inmunología
10.
MAbs ; 10(4): 651-663, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29553870

RESUMEN

Plasmacytoid dendritic cells (pDCs) play a central role for both innate and adaptive antiviral responses, as they direct immune responses through their unique ability to produce substantial concentrations of type I interferon (IFNs) upon viral encounter while also activating multiple immune cells, including macrophages, DCs, B, natural killer and T cells. Recent evidence clearly indicates that pDCs also play a crucial role in some cancers and several auto-immune diseases. Although treatments are currently available to patients with such pathologies, many are not fully efficient. We are proposing here, as a new targeted-based therapy, a novel chimeric monoclonal antibody (mAb) that mediates a strong cellular cytotoxicity directed against a specific human pDC marker, CD303. This antibody, ch122A2 mAb, is characterized by low fucose content in its human IgG1 constant (Fc) region, which induces strong in vitro and in vivo activity against human pDCs. We demonstrated that this effect relates in part to its specific Fc region glycosylation pattern, which increased affinity for CD16/FcγRIIIa. Importantly, ch122A2 mAb induces the down-modulation of CpG-induced IFN-α secretion by pDCs. Additionally, ch122A2 mAb shows in vitro high pDC depletion mediated by antibody-dependent cell-mediated cytotoxicity and antibody-dependent cellular phagocytosis. Remarkably, in vivo ch122A2 mAb efficacy is also demonstrated in humanized mice, resulting in significant pDC depletion in bloodstream and secondary lymphoid organs such as spleen. Together, our data indicates that ch122A2 mAb could represent a promising cytotoxic mAb candidate for pathologies in which decreasing type I IFNs or pDCs depleting may improve patient prognosis.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células Dendríticas , Lectinas Tipo C/antagonistas & inhibidores , Glicoproteínas de Membrana/antagonistas & inhibidores , Receptores Inmunológicos/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Ratones , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología
11.
Toxins (Basel) ; 9(10)2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28974033

RESUMEN

The goal of the AntiBotABE Program was the development of recombinant antibodies that neutralize botulinum neurotoxins (BoNT) A, B and E. These serotypes are lethal and responsible for most human botulinum cases. To improve therapeutic efficacy, the heavy and light chains (HC and LC) of the three BoNT serotypes were targeted to achieve a synergistic effect (oligoclonal antibodies). For antibody isolation, macaques were immunized with the recombinant and non-toxic BoNT/A, B or E, HC or LC, followed by the generation of immune phage-display libraries. Antibodies were selected from these libraries against the holotoxin and further analyzed in in vitro and ex vivo assays. For each library, the best ex vivo neutralizing antibody fragments were germline-humanized and expressed as immunoglobulin G (IgGs). The IgGs were tested in vivo, in a standardized model of protection, and challenged with toxins obtained from collections of Clostridium strains. Protective antibody combinations against BoNT/A and BoNT/B were evidenced and for BoNT/E, the anti-LC antibody alone was found highly protective. The combination of these five antibodies as an oligoclonal antibody cocktail can be clinically and regulatorily developed while their high "humanness" predicts a high tolerance in humans.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Toxinas Botulínicas/inmunología , Neurotoxinas/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Humanos , Inmunización , Proteínas Recombinantes/inmunología
12.
Oncotarget ; 8(23): 37061-37079, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28427157

RESUMEN

Ovarian cancer is the leading cause of death in women with gynecological cancers and despite recent advances, new and more efficient therapies are crucially needed. Müllerian Inhibiting Substance type II Receptor (MISRII, also named AMHRII) is expressed in most ovarian cancer subtypes and is a novel potential target for ovarian cancer immunotherapy. We previously developed and tested 12G4, the first murine monoclonal antibody (MAb) against human MISRII. Here, we report the humanization, affinity maturation and glyco-engineering steps of 12G4 to generate the Fc-optimized 3C23K MAb, and the evaluation of its in vivo anti-tumor activity. The epitopes of 3C23K and 12G4 were strictly identical and 3C23K affinity for MISRII was enhanced by a factor of about 14 (KD = 5.5 × 10-11 M vs 7.9 × 10-10 M), while the use of the EMABling® platform allowed the production of a low-fucosylated 3C23K antibody with a 30-fold KD improvement of its affinity to FcγRIIIa. In COV434-MISRII tumor-bearing mice, 3C23K reduced tumor growth more efficiently than 12G4 and its combination with carboplatin was more efficient than each monotherapy with a mean tumor size of 500, 1100 and 100 mm3 at the end of treatment with 3C23K (10 mg/kg, Q3-4D12), carboplatin (60 mg/kg, Q7D4) and 3C23K+carboplatin, respectively. Conversely, 3C23K-FcKO, a 3C23K form without affinity for the FcγRIIIa receptor, did not display any anti-tumor effect in vivo. These results strongly suggested that 3C23K mechanisms of action are mainly Fc-related. In vitro, antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP) were induced by 3C23K, as demonstrated with human effector cells. Using human NK cells, 50% of the maximal lysis was obtained with a 46-fold lower concentration of low-fucosylated 3C23K (2.9 ng/ml) than of 3C23K expressed in CHO cells (133.35 ng/ml). As 3C23K induced strong ADCC with human PBMC but almost none with murine PBMC, antibody-dependent cell phagocytosis (ADCP) was then investigated. 3C23K-dependent (100 ng/ml) ADCP was more active with murine than human macrophages (only 10% of living target cells vs. about 25%). These in vitro results suggest that the reduced ADCC with murine effectors could be partially balanced by ADCP activity in in vivo experiments. Taken together, these preclinical data indicate that 3C23K is a new promising therapeutic candidate for ovarian cancer immunotherapy and justify its recent introduction in a phase I clinical trial.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Receptores de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Femenino , Glicosilación , Humanos , Ratones Desnudos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Ingeniería de Proteínas
13.
FEBS Lett ; 580(26): 6206-10, 2006 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-17069809

RESUMEN

We investigated parabutoporin (PP), an antimicrobial scorpion peptide, to understand its inhibition on NADPH oxidase in human PMN. We show that PP is a good substrate for all PKC-isotypes, implicated in the activation of NADPH oxidase, and acts as a potent competitive inhibitor of in vitro p47(phox)-phosphorylation by PKC-alpha, -betaI, -betaII and -delta, but not PKC-zeta. In PMN, PP also inhibits the PMA-stimulated phosphorylation of p47(phox) and its subsequent translocation. In contrast, PP affects the PKC-independent activation to a much lesser degree. This indicates that PP inhibits the activation of NADPH oxidase at submicromolar concentrations in a strongly PKC-dependent manner.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , NADPH Oxidasas/antagonistas & inhibidores , Neutrófilos/efectos de los fármacos , Proteína Quinasa C/metabolismo , Venenos de Escorpión/química , Péptidos Catiónicos Antimicrobianos/metabolismo , Unión Competitiva , Humanos , NADPH Oxidasas/metabolismo , Fosforilación , Isoformas de Proteínas , Venenos de Escorpión/metabolismo , Venenos de Escorpión/farmacología
14.
Thromb Haemost ; 96(5): 671-84, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17080226

RESUMEN

Fab-fragments of the monoclonal antibody 6B4, raised against human glycoprotein Ibalpha (GPIbalpha), have a powerful antithrombotic effect in baboons by blocking the GPIbalpha binding site for von Willebrand factor (VWF), without significant prolongation of the skin bleeding time. In order to bring this antibody to the clinic,we here humanized for the first time an anti-human GPIbalpha by variable-domain resurfacing guided by computer modeling. First, the genes coding for the variable regions of the heavy and light chains of 6B4 were cloned and sequenced. Based on this, a three-dimensional structure of the Fv-fragment was constructed by using homology-based modeling, and with this and comparison with antibodies with known structure,"murine" putative immunogenic residues which are exposed, were changed for "human-like" residues. The humanized Fab-fragment, h6B4-Fab, was constructed in the pKaneo vector system, expressed and purified and showed in vitro an unaltered, even slightly higher binding affinity for its antigen than the murine form as determined by different ELISA set-ups and surface plasmon resonance. Finally, injection of doses of 0.1 to 1.5 mg/kg of h6B4-Fab in baboons showed that both pharmacokinetics and ex-vivo bio-activity of the molecule were to a large extent preserved. In conclusion,the method used here to humanize 6B4 by resurfacing resulted in a fully active derivative, which is now ready for further development.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Diseño de Fármacos , Fibrinolíticos/farmacocinética , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/inmunología , Animales , Anticuerpos Monoclonales/genética , Clonación Molecular , Simulación por Computador , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Ratones , Modelos Moleculares , Papio , Conformación Proteica , Ingeniería de Proteínas
15.
Toxins (Basel) ; 8(9)2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27626446

RESUMEN

Botulinum neurotoxins (BoNTs) cause botulism and are the deadliest naturally-occurring substances known to humans. BoNTs have been classified as one of the category A agents by the Centers for Disease Control and Prevention, indicating their potential use as bioweapons. To counter bio-threat and naturally-occurring botulism cases, well-tolerated antibodies by humans that neutralize BoNTs are relevant. In our previous work, we showed the neutralizing potential of macaque (Macaca fascicularis)-derived scFv-Fc (scFv-Fc ELC18) by in vitro endopeptidase immunoassay and ex vivo mouse phrenic nerve-hemidiaphragm assay by targeting the light chain of the botulinum neurotoxin type E (BoNT/E). In the present study, we germline-humanized scFv-Fc ELC18 into a full IgG hu8ELC18 to increase its immunotolerance by humans. We demonstrated the protection and prophylaxis capacity of hu8ELC18 against BoNT/E in a mouse model. A concentration of 2.5 ng/mouse of hu8ELC18 protected against 5 mouse lethal dose (MLD) in a mouse protection assay and complete neutralization of 1 LD50 of pure BoNT/E toxin was achieved with 8 ng of hu8ELC18 in mouse paralysis assay. Furthermore, hu8ELC18 protected mice from 5 MLD if injected up to 14 days prior to intraperitoneal BoNT/E administration. This newly-developed humanized IgG is expected to have high tolerance in humans.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Antídotos/farmacología , Antitoxinas/farmacología , Toxinas Botulínicas/antagonistas & inhibidores , Botulismo/prevención & control , Clostridium botulinum/efectos de los fármacos , Anticuerpos de Cadena Única/farmacología , Animales , Toxinas Botulínicas/inmunología , Botulismo/inmunología , Botulismo/microbiología , Clostridium botulinum/inmunología , Clostridium botulinum/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones
16.
PLoS One ; 11(8): e0161446, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27560688

RESUMEN

Botulinum neurotoxins (BoNTs) are counted among the most toxic substances known and are responsible for human botulism, a life-threatening disease characterized by flaccid muscle paralysis that occurs naturally by food poisoning or colonization of the gastrointestinal tract by BoNT-producing clostridia. To date, 7 serologically distinct serotypes of BoNT (serotype A-G) are known. Due to the high toxicity of BoNTs the Centers for Disease Control and Prevention (CDC) have classified BoNTs as category A agent, including the six biological agents with the highest potential risk of use as bioweapons. Well tolerated antibodies neutralizing BoNTs are required to deal with the potential risk. In a previous work, we described the development of scFv and scFv-Fc (Yumab) from macaque origin (Macaca fascicularis) neutralizing BoNT/A and B by targeting the heavy and light chain of each serotype. In the present study, we humanized the macaque antibodies SEM120-IIIC1 (anti-BoNT/A light chain), A1HC38 (anti-BoNT/A heavy chain), BLC3 (anti-BoNT/B light chain) and B2-7 (anti-BoNT/B heavy chain) by germline-humanization to obtain a better potential immunotolerance in humans. We increased the Germinality Index (GI) of SEM120-IIIC1 to 94.5%, for A1HC38, to 95% for BLC3 and to 94.4% for B2-7. Furthermore, the neutralization efficacies of the germline-humanized antibodies were analyzed in lethal and non-lethal in vivo mouse assays as full IgG. The germline-humanized IgGs hu8SEM120-IIIC1, hu8A1HC38, hu8BLC3 and hu8B2-7 were protective in vivo, when anti-heavy and anti-light chain antibodies were combined. The synergistic effect and high humanness of the selected IgGs makes them promising lead candidates for further clinical development.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Toxinas Botulínicas Tipo A/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Botulismo/inmunología , Clostridium botulinum , Femenino , Humanos , Inmunoglobulina G/inmunología , Macaca fascicularis/inmunología , Ratones , Pruebas de Neutralización , Anticuerpos de Cadena Única/inmunología
17.
J Control Release ; 234: 21-32, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27173943

RESUMEN

The high toxicity of ricin and its ease of production have made it a major bioterrorism threat worldwide. There is however no efficient and approved treatment for poisoning by ricin inhalation, although there have been major improvements in diagnosis and therapeutic strategies. We describe the development of an anti-ricin neutralizing monoclonal antibody (IgG 43RCA-G1) and a device for its rapid and effective delivery into the lungs for an application in humans. The antibody is a full-length IgG and binds to the ricin A-chain subunit with a high affinity (KD=53pM). Local administration of the antibody into the respiratory tract of mice 6h after pulmonary ricin intoxication allowed the rescue of 100% of intoxicated animals. Specific operational constraints and aerosolization stresses, resulting in protein aggregation and loss of activity, were overcome by formulating the drug as a dry-powder that is solubilized extemporaneously in a stabilizing solution to be nebulized. Inhalation studies in mice showed that this formulation of IgG 43RCA-G1 did not induce pulmonary inflammation. A mesh nebulizer was customized to improve IgG 43RCA-G1 deposition into the alveolar region of human lungs, where ricin aerosol particles mostly accumulate. The drug delivery system also comprises a semi-automatic reconstitution system to facilitate its use and a specific holding chamber to maximize aerosol delivery deep into the lung. In vivo studies in monkeys showed that drug delivery with the device resulted in a high concentration of IgG 43RCA-G1 in the airways for at least 6h after local deposition, which is consistent with the therapeutic window and limited passage into the bloodstream.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Sustancias para la Guerra Química/envenenamiento , Sistemas de Liberación de Medicamentos/métodos , Lesión Pulmonar/tratamiento farmacológico , Alveolos Pulmonares/efectos de los fármacos , Ricina/envenenamiento , Aerosoles , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/toxicidad , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Neutralizantes/toxicidad , Femenino , Humanos , Células Jurkat , Lesión Pulmonar/inducido químicamente , Macaca fascicularis , Masculino , Ratones Endogámicos BALB C , Nebulizadores y Vaporizadores , Distribución Tisular
18.
Front Immunol ; 6: 39, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25699055

RESUMEN

Despite the reasonably long half-life of immunoglogulin G (IgGs), market pressure for higher patient convenience while conserving efficacy continues to drive IgG half-life improvement. IgG half-life is dependent on the neonatal Fc receptor (FcRn), which among other functions, protects IgG from catabolism. FcRn binds the Fc domain of IgG at an acidic pH ensuring that endocytosed IgG will not be degraded in lysosomal compartments and will then be released into the bloodstream. Consistent with this mechanism of action, several Fc-engineered IgG with increased FcRn affinity and conserved pH dependency were designed and resulted in longer half-life in vivo in human FcRn-transgenic mice (hFcRn), cynomolgus monkeys, and recently in healthy humans. These IgG variants were usually obtained by in silico approaches or directed mutagenesis in the FcRn-binding site. Using random mutagenesis, combined with a pH-dependent phage display selection process, we isolated IgG variants with improved FcRn-binding, which exhibited longer in vivo half-life in hFcRn mice. Interestingly, many mutations enhancing Fc/FcRn interaction were located at a distance from the FcRn-binding site validating our random molecular approach. Directed mutagenesis was then applied to generate new variants to further characterize our IgG variants and the effect of the mutations selected. Since these mutations are distributed over the whole Fc sequence, binding to other Fc effectors, such as complement C1q and FcγRs, was dramatically modified, even by mutations distant from these effectors' binding sites. Hence, we obtained numerous IgG variants with increased FcRn-binding and different binding patterns to other Fc effectors, including variants without any effector function, providing distinct "fit-for-purpose" Fc molecules. We therefore provide evidence that half-life and effector functions should be optimized simultaneously as mutations can have unexpected effects on all Fc receptors that are critical for IgG therapeutic efficacy.

19.
MAbs ; 6(2): 422-36, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24492301

RESUMEN

While glyco-engineered monoclonal antibodies (mAbs) with improved antibody-dependent cell-mediated cytotoxicity (ADCC) are reaching the market, extensive efforts have also been made to improve their pharmacokinetic properties to generate biologically superior molecules. Most therapeutic mAbs are human or humanized IgG molecules whose half-life is dependent on the neonatal Fc receptor FcRn. FcRn reduces IgG catabolism by binding to the Fc domain of endocytosed IgG in acidic lysosomal compartments, allowing them to be recycled into the blood. Fc-engineered mAbs with increased FcRn affinity resulted in longer in vivo half-life in animal models, but also in healthy humans. These Fc-engineered mAbs were obtained by alanine scanning, directed mutagenesis or in silico approach of the FcRn binding site. In our approach, we applied a random mutagenesis technology (MutaGen™) to generate mutations evenly distributed over the whole Fc sequence of human IgG1. IgG variants with improved FcRn-binding were then isolated from these Fc-libraries using a pH-dependent phage display selection process. Two successive rounds of mutagenesis and selection were performed to identify several mutations that dramatically improve FcRn binding. Notably, many of these mutations were unpredictable by rational design as they were located distantly from the FcRn binding site, validating our random molecular approach. When produced on the EMABling(®) platform allowing effector function increase, our IgG variants retained both higher ADCC and higher FcRn binding. Moreover, these IgG variants exhibited longer half-life in human FcRn transgenic mice. These results clearly demonstrate that glyco-engineering to improve cytotoxicity and protein-engineering to increase half-life can be combined to further optimize therapeutic mAbs.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina G/metabolismo , Inmunoterapia/métodos , Ingeniería de Proteínas/métodos , Receptores Fc/metabolismo , Animales , Anticuerpos Monoclonales/genética , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Técnicas de Visualización de Superficie Celular , Citotoxicidad Inmunológica/genética , Glicosilación , Semivida , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Inmunoglobulina G/genética , Inmunoterapia/tendencias , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Mutación/genética , Receptores Fc/genética , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/inmunología , Receptores de IgG/metabolismo
20.
Thromb Haemost ; 104(2): 392-401, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20508902

RESUMEN

Platelets play an essential role in the development of cardiovascular diseases and are the target of several agents that can inhibit their function. Despite the existence of a wide array of techniques to study platelet function, an assay to evaluate several platelet signalling pathways in a high-throughput fashion, combined with minimal blood volume and handling is still needed. We have developed a sensitive assay in the form of a sandwich ELISA where monoclonal antibodies against P-selectin or alphaIIbbeta3 and GPIbalpha were used to capture and detect platelets, respectively, in the presence of five different agonists [ADP, TRAP (thrombin receptor agonist), U46619 (thromboxane A2 analogue), collagen-related-peptide, and arachidonic acid]. Binding of platelets to the antibodies increased dose-dependently with the concentration of either agonist, while binding of ADP-activated platelets was abrogated when inhibitors of platelet activation were concomitantly added. The test showed good sample reproducibility in 15 healthy donors with conserved platelet response to agonists throughout the assay. Healthy subjects could be identified as normal-, hypo- or hyper-responders for each agonist, which for most cases (73%) was confirmed upon retesting. Finally, we demonstrated that the platelet ELISA assay can not only be used in platelet-rich plasma but also in whole blood; it now awaits large scale studies to assess its full screening and diagnostic values.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática , Ensayos Analíticos de Alto Rendimiento , Activación Plaquetaria , Pruebas de Función Plaquetaria , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Adenosina Difosfato , Alprostadil/farmacología , Anticuerpos Monoclonales , Ácido Araquidónico , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Plaquetas/fisiología , Proteínas Portadoras , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Glicoproteínas/inmunología , Humanos , Inmunoglobulinas/inmunología , Integrina alfa2/inmunología , Integrina beta3/inmunología , Variaciones Dependientes del Observador , Selectina-P/inmunología , Péptidos , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , S-Nitroso-N-Acetilpenicilamina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA