Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Antiviral Res ; : 105952, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38945484

RESUMEN

Argentine hemorrhagic fever, caused by Junín virus (JUNV), is the most common of the South American arenaviral hemorrhagic fevers. The disease has a case fatality rate of 15-30% in untreated patients. Although early intervention with immune plasma is effective, diminishing stocks and limited availability outside of Argentina underscores the need for new therapeutics. Ideally, these would be broadly active agents effective against all the pathogenic arenaviruses. The fusion inhibitor LHF-535 and the nucleoside analog favipiravir have shown promise in animal models of Lassa fever, a disease endemic in parts of Africa and the most prominent of the arenaviral hemorrhagic fevers. Against JUNV, a high dose of favipiravir is required to achieve protection in the gold-standard guinea pig infection model. Here, we demonstrate a synergistic effect by the coadministration of LHF-535 with a sub-optimal dose of favipiravir in guinea pigs challenged with JUNV. Administered individually, LHF-535 and sub-optimal favipiravir only delayed the onset of severe disease. However, combined dosing of the drugs afforded complete protection against lethal JUNV infection in guinea pigs. The benefits of the drug combination were also evident by the absence of viremia and infectious virus in tissues compared to guinea pigs treated with only the placebos. Thus, combined targeting of JUNV-endosomal membrane fusion and the viral polymerase with pan-arenaviral LHF-535 and favipiravir may expand their indication beyond Lassa fever, providing a significant barrier to drug resistance.

3.
Front Immunol ; 14: 1311658, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38152397

RESUMEN

Background: Immune checkpoint therapies have led to significant breakthroughs in cancer patient treatment in recent years. However, their efficiency is variable, and resistance to immunotherapies is common. VISTA is an immune-suppressive checkpoint inhibitor of T cell response belonging to the B7 family and a promising novel therapeutic target. VISTA is expressed in the immuno-suppressive tumor microenvironment, primarily by myeloid lineage cells, and its genetic knockout or antibody blockade restores an efficient antitumor immune response. Methods: Fully human monoclonal antibodies directed against VISTA were produced after immunizing humanized Trianni mice and single B cell sequencing. Anti-VISTA antibodies were evaluated for specificity, cross-reactivity, monocyte and T cell activation, Fc-effector functions, and antitumor efficacy using in vitro and in vivo models to select the KVA12123 antibody lead candidate. The pharmacokinetics and safety profiles of KVA12123 were evaluated in cynomolgus monkeys. Results: Here, we report the development of a clinical candidate anti-VISTA monoclonal antibody, KVA12123. KVA12123 showed high affinity binding to VISTA through a unique epitope distinct from other clinical-stage anti-VISTA monoclonal antibodies. This clinical candidate demonstrated high specificity against VISTA with no cross-reactivity detected against other members of the B7 family. KVA12123 blocked VISTA binding to its binding partners. KVA12123 induced T cell activation and demonstrated NK-mediated monocyte activation. KVA12123 treatment mediated strong single-agent antitumor activity in several syngeneic tumor models and showed enhanced efficacy in combination with anti-PD-1 treatment. This clinical candidate was engineered to improve its pharmacokinetic characteristics and reduce Fc-effector functions. It was well-tolerated in preclinical toxicology studies in cynomolgus monkeys, where hematology, clinical chemistry evaluations, and clinical observations revealed no indicators of toxicity. No cytokines associated with cytokine release syndrome were elevated. Conclusion: These results establish that KVA12123 is a promising drug candidate with a distinct but complementary mechanism of action of the first generation of immune checkpoint inhibitors. This antibody is currently evaluated alone and in combination with pembrolizumab in a Phase 1/2 open-label clinical trial in patients with advanced solid tumors.


Asunto(s)
Anticuerpos Monoclonales , Inhibidores de Puntos de Control Inmunológico , Neoplasias , Animales , Humanos , Ratones , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Macaca fascicularis , Neoplasias/terapia , Linfocitos T , Microambiente Tumoral
4.
Sci Rep ; 12(1): 19911, 2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36402782

RESUMEN

LHF-535 is a small molecule antiviral currently in development for the treatment of Lassa fever, a zoonotic disease endemic in West Africa that generates significant morbidity and mortality. Current treatment options are inadequate, and there are no approved therapeutics or vaccines for Lassa fever. LHF-535 was evaluated in a lethal guinea pig model of Lassa pathogenesis, using once-daily administration of a fixed dose (50 mg/kg/day) initiating either 1 or 3 days after inoculation with a lethal dose of Lassa virus. LHF-535 reduced viremia and clinical signs and protected all animals from lethality. A subset of surviving animals was rechallenged four months later with a second lethal challenge of Lassa virus and were found to be protected from disease. LHF-535 pharmacokinetics at the protective dose in guinea pigs showed plasma concentrations well within the range observed in clinical trials in healthy volunteers, supporting the continued development of LHF-535 as a Lassa therapeutic.


Asunto(s)
Fiebre de Lassa , Cobayas , Animales , Fiebre de Lassa/tratamiento farmacológico , Fiebre de Lassa/prevención & control , Antivirales/farmacología , Antivirales/uso terapéutico , Virus Lassa , Viremia/tratamiento farmacológico , Vacunación
5.
PLoS One ; 7(8): e43332, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22912856

RESUMEN

Chemokines play a key role in leukocyte recruitment during inflammation and are implicated in the pathogenesis of a number of autoimmune diseases. As such, inhibiting chemokine signaling has been of keen interest for the development of therapeutic agents. This endeavor, however, has been hampered due to complexities in the chemokine system. Many chemokines have been shown to signal through multiple receptors and, conversely, most chemokine receptors bind to more than one chemokine. One approach to overcoming this complexity is to develop a single therapeutic agent that binds and inactivates multiple chemokines, similar to an immune evasion strategy utilized by a number of viruses. Here, we describe the development and characterization of a novel therapeutic antibody that targets a subset of human CC chemokines, specifically CCL3, CCL4, and CCL5, involved in chronic inflammatory diseases. Using a sequential immunization approach, followed by humanization and phage display affinity maturation, a therapeutic antibody was developed that displays high binding affinity towards the three targeted chemokines. In vitro, this antibody potently inhibits chemotaxis and chemokine-mediated signaling through CCR1 and CCR5, primary chemokine receptors for the targeted chemokines. Furthermore, we have demonstrated in vivo efficacy of the antibody in a SCID-hu mouse model of skin leukocyte migration, thus confirming its potential as a novel therapeutic chemokine antagonist. We anticipate that this antibody will have broad therapeutic utility in the treatment of a number of autoimmune diseases due to its ability to simultaneously neutralize multiple chemokines implicated in disease pathogenesis.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Quimiocinas CC/inmunología , Inmunomodulación/inmunología , Inmunoterapia/métodos , Transducción de Señal/inmunología , Animales , Anticuerpos Neutralizantes/uso terapéutico , Enfermedades Autoinmunes/inmunología , Quimiotaxis/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Fosforilación , Resonancia por Plasmón de Superficie
6.
Mol Ther ; 15(2): 264-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17235303

RESUMEN

We previously reported that administration of an adeno-associated virus 2 (AAV2) vector encoding a rat tumor necrosis factor (TNF) receptor-immunoglobulin Fc (TNFR:Fc) fusion gene to rats with streptococcal cell wall-induced arthritis resulted in suppression of joint inflammation and cartilage and bone destruction, as well as expression of joint proinflammatory cytokines. In this study, we used an alternate rat model of arthritis to compare the serum levels and duration of TNFR:Fc protein expression following intramuscular administration of pseudotyped AAV-TNFR:Fc vectors based on serotypes 1, 2, and 5. All three pseudotyped AAV-TNFR:Fc vectors led to sustained expression of serum TNFR:Fc protein for at least one year. Serum TNFR:Fc protein levels in rats administered intramuscularly with AAV2/1-TNFR:Fc vector were up to 100- and 10-fold higher than in rats administered the AAV2-TNFR:Fc or AAV2/5-TNFR:Fc vectors, respectively. A single intramuscular administration of AAV2/1-TNFR:Fc vector at vector doses ranging from 10(10) to 10(12) DNase-resistant particles (DRP) per animal, resulted in complete and long-term suppression of recurrent joint inflammation for at least 150 days. Our results establish a proof of concept for administration of an AAV2/1-TNFR:Fc vector to the muscle to achieve long-term, sustained and therapeutically relevant levels of TNFR:Fc protein to treat chronic systemic inflammatory joint diseases.


Asunto(s)
Artritis Experimental/terapia , Terapia Genética/métodos , Fragmentos Fc de Inmunoglobulinas/genética , Receptores del Factor de Necrosis Tumoral/genética , Virus Satélites/genética , Animales , Artritis Experimental/sangre , Bovinos , Línea Celular , Femenino , Vectores Genéticos/genética , Células HeLa , Humanos , Inyecciones Intramusculares , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/genética
7.
Brain Behav Evol ; 65(4): 278-92, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15761219

RESUMEN

Thalamocortical connections are a neuroanatomical feature shared among vertebrates, although the extent and organization of these connections vary among species. From an evolutionary standpoint, reptiles represent early stages of the pattern of connectivity between the thalamus and cortex, and elucidation of these pathways may help to reveal the biological significance of these projections. The present tract tracing study was performed to examine the organization of thalamocortical projections in the pond turtle, Pseudemys scripta elegans. All experiments were carried out using in vitro brain preparations. Injections of neurobiotin into the medial cortex resulted in labeled neurons in the ipsilateral dorsomedial anterior nucleus of the thalamus, those in the dorsomedial cortex labeled neurons in the dorsolateral anterior nucleus, and injections into the dorsal cortex resulted in labeled neurons in the dorsal lateral geniculate nucleus of the thalamus. Injections of neurobiotin into these thalamic nuclei confirmed the projections to the cortex. Finally, neurobiotin injections primarily into the medial cortex resulted in bilateral label of axons and terminals in the suprapeduncular nucleus of the hypothalamus. The results of the neurobiotin injections revealed a topographic pattern of thalamocortical connections such that medial cortical regions connect with medial thalamic nuclei and lateral cortical regions connect with lateral nuclei. These findings suggest that the presence of functionally segregated thalamocortical projections is a conserved feature of brain organization among amniotes. Moreover, this work describes a descending pathway linking cortical regions with the red nucleus via the hypothalamus thereby providing indirect cortical control of the reptilian rubrospinal system.


Asunto(s)
Mapeo Encefálico , Corteza Cerebral/citología , Vías Nerviosas/citología , Tálamo/citología , Tortugas/anatomía & histología , Animales , Núcleo Rojo/citología , Núcleos Talámicos/citología
8.
J Virol ; 78(22): 12355-65, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15507622

RESUMEN

This study evaluated and compared delivery of the tumor necrosis factor alpha receptor (TNFR)-immunoglobulin G1 (IgG1) Fc fusion (TNFR:Fc) gene to the lung by single and repeat administrations of multiple pseudotyped adeno-associated virus (AAV) vectors as a means for achieving systemic distribution of the soluble TNFR:Fc protein. A single endotracheal administration of AAV[2/5]cytomegalovirus (CMV)-TNFR:Fc vector (containing the AAV2 inverted terminal repeats and AAV5 capsid) to the rat lung resulted in long-term, high levels of serum TNFR:Fc protein that gradually declined over a period of 8 months. Endotracheal delivery of AAV[2/1]CMV-TNFR:Fc resulted in serum TNFR:Fc protein levels that were detectable for at least 4 months but were 10-fold lower than that of the AAV[2/5] vector. In contrast, secretion of the TNFR:Fc protein following pulmonary delivery of AAV[2/2]CMV-TNFR:Fc vector was very inefficient, and the protein was detected in the blood only when an airway epithelial cell-specific promoter, CC10, was substituted for the CMV enhancer/promoter to control transgene expression. In the context of AAV[2/5], the CC10 promoter was as efficient as CMV enhancer/promoter in generating similar levels of systemic TNFR:Fc protein, suggesting that this protein is secreted primarily from the airway epithelium. In mice, comparable long-term secretion of TNFR:Fc protein was demonstrated after AAV[2/2] and AAV[2/5] delivery, although the kinetics of transduction appeared to be different. All pseudotyped AAV vectors elicited serum anti-AAV capsid-neutralizing antibody responses, but these did not prevent lung transduction and efficient secretion of TNFR:Fc protein to the circulation following readministration with AAV[2/5]. These results highlight the potential utility of AAV vectors containing serotype 5 capsid to deliver and redeliver genes of secreted proteins to the lung to achieve long-term systemic protein expression.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Fragmentos Fc de Inmunoglobulinas/biosíntesis , Pulmón/metabolismo , Receptores del Factor de Necrosis Tumoral/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Animales , Anticuerpos Antivirales/sangre , Línea Celular , Dependovirus/inmunología , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/sangre , Ratas , Ratas Endogámicas Lew , Receptores del Factor de Necrosis Tumoral/sangre , Proteínas Recombinantes de Fusión/sangre , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA