Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Vet Immunol Immunopathol ; 258: 110574, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36842258

RESUMEN

Lokivetmab (Cytopoint®, Zoetis) is a canine monoclonal antibody that specifically binds and neutralizes interleukin (IL)-31. Lokivetmab is approved for use in dogs for the treatment of atopic dermatitis (AD) and allergic dermatitis. The laboratory safety of lokivetmab was evaluated in 2 studies by adapting the science-based, case-by-case approach used for preclinical and early clinical safety evaluation of human biopharmaceuticals. The main objectives were to demonstrate the safety of lokivetmab in healthy laboratory Beagle dogs by using integrated clinical, morphologic, and functional evaluations. In Study 1, dogs were treated s.c. with saline or lokivetmab at 3.3 mg/kg (1X, label dose) or 10 mg/kg (3X intended dose) for 7 consecutive monthly doses, with terminal pathology and histology assessments. In Study 2, the functional immune response was demonstrated in naïve dogs using the T-cell dependent antibody response (TDAR) test with 2 different dose levels of unadjuvanted keyhole limpet hemocyanin (KLH) as the model immunogen. The primary endpoint was anti-KLH IgG antibody titer, and secondary endpoints were ex vivo IL-2 enzyme-linked immunospot (ELISpot) and peripheral blood mononuclear cell lymphoproliferation assays. Both studies included monitoring general health, periodic veterinary clinical evaluations, serial clinical pathology and toxicokinetics, and monitoring for anti-drug antibodies. In both studies, the health of dogs receiving lokivetmab was similar to controls, with no treatment-related changes uncovered. Extensive pathology evaluations of immune tissues (Study 1) revealed no lokivetmab-related morphologic changes, and in dogs treated at 10 mg/kg lokivetmab, immunization with the model antigen KLH did not impair the functional antibody or T-cell recall responses. There were no immunogenicity-related or hypersensitivity-related responses observed in either study. These studies in healthy laboratory dogs showed that lokivetmab was well-tolerated, did not produce any treatment-related effects, and had no effect on immune system morphology or its functional response. These studies also demonstrated the utility of a science-based case-by-case approach to the safety evaluation of a veterinary biopharmaceutical product.


Asunto(s)
Dermatitis Atópica , Enfermedades de los Perros , Animales , Perros , Humanos , Anticuerpos Monoclonales , Formación de Anticuerpos , Dermatitis Atópica/veterinaria , Enfermedades de los Perros/tratamiento farmacológico , Hemocianinas/farmacología , Hemocianinas/uso terapéutico , Leucocitos Mononucleares , Linfocitos T , Interleucinas
2.
PLoS One ; 12(1): e0170504, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28107431

RESUMEN

Lack of safe and effective adjuvants is a major hindrance to the development of efficacious vaccines. Signaling via CD40 pathway leads to enhanced antigen processing and presentation, nitric oxide expression, pro-inflammatory cytokine expression by antigen presenting cells, and stimulation of B-cells to undergo somatic hypermutation, immunoglobulin class switching, and proliferation. Agonistic anti-CD40 antibodies have shown promising adjuvant qualities in human and mouse vaccine studies. An anti-CD40 monoclonal antibody (mAb), designated 2E4E4, was identified and shown to have strong agonistic effects on primary cells from multiple livestock species. The mAb recognize swine, bovine, caprine, and ovine CD40, and evoked 25-fold or greater proliferation of peripheral blood mononuclear cells (PBMCs) from these species relative to cells incubated with an isotype control (p<0.001). In addition, the mAb induced significant nitric oxide (p<0.0001) release by bovine macrophages. Furthermore, the mAb upregulated the expression of MHC-II by PBMCs, and stimulated significant (p<0.0001) IL-1α, IL6, IL-8, and TNF-α expression by PBMCs. These results suggest that the mAb 2E4E4 can target and stimulate cells from multiple livestock species and thus, it is a potential candidate for adjuvant development. This is the first study to report an anti-swine CD40 agonistic mAb that is also broadly reactive against multiple species.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Anticuerpos Monoclonales/inmunología , Antígenos CD40/inmunología , Animales , Bovinos/inmunología , Reacciones Cruzadas/inmunología , Citocinas/metabolismo , Femenino , Citometría de Flujo , Genes MHC Clase II/inmunología , Cabras/inmunología , Células HEK293 , Humanos , Leucocitos Mononucleares/inmunología , Ratones , Ratones Endogámicos BALB C , Ovinos/inmunología , Porcinos/inmunología
3.
Clin Vaccine Immunol ; 23(11): 888-900, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27628166

RESUMEN

The African swine fever virus (ASFV) causes a fatal hemorrhagic disease in domestic swine, and at present no treatment or vaccine is available. Natural and gene-deleted, live attenuated strains protect against closely related virulent strains; however, they are yet to be deployed and evaluated in the field to rule out chronic persistence and a potential for reversion to virulence. Previous studies suggest that antibodies play a role in protection, but induction of cytotoxic T lymphocytes (CTLs) could be the key to complete protection. Hence, generation of an efficacious subunit vaccine depends on identification of CTL targets along with a suitable delivery method that will elicit effector CTLs capable of eliminating ASFV-infected host cells and confer long-term protection. To this end, we evaluated the safety and immunogenicity of an adenovirus-vectored ASFV (Ad-ASFV) multiantigen cocktail formulated in two different adjuvants and at two immunizing doses in swine. Immunization with the cocktail rapidly induced unprecedented ASFV antigen-specific antibody and cellular immune responses against all of the antigens. The robust antibody responses underwent rapid isotype switching within 1 week postpriming, steadily increased over a 2-month period, and underwent rapid recall upon boost. Importantly, the primed antibodies strongly recognized the parental ASFV (Georgia 2007/1) by indirect fluorescence antibody (IFA) assay and Western blotting. Significant antigen-specific gamma interferon-positive (IFN-γ+) responses were detected postpriming and postboosting. Furthermore, this study is the first to demonstrate induction of ASFV antigen-specific CTL responses in commercial swine using Ad-ASFV multiantigens. The relevance of the induced immune responses in regard to protection needs to be evaluated in a challenge study.


Asunto(s)
Virus de la Fiebre Porcina Africana/inmunología , Antígenos Virales/inmunología , Inmunidad Celular , Inmunogenicidad Vacunal , Vacunas Virales/inmunología , Adenoviridae/genética , Animales , Antígenos Virales/química , Vectores Genéticos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Porcinos , Linfocitos T Citotóxicos/inmunología , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/inmunología , Vacunas Virales/efectos adversos , Virulencia
4.
Vet Immunol Immunopathol ; 153(1-2): 128-33, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23351642

RESUMEN

Hydrocortisone is widely accepted as an anti-inflammatory agent and there are many products available containing hydrocortisone as an active ingredient. Surprisingly, there is little data available specifically on the immunological effects of hydrocortisone in large animals. Glucocorticoids are well-characterized for their ability to repress inflammation via a wide variety of mechanisms including suppression of cytokine production. In this study the effects of hydrocortisone on IFN-γ production by equine, bovine, and ovine PBMCs were assessed using flow cytometric or ELISpot analysis. Hydrocortisone suppressed mitogen-driven IFN-γ production by PBMCs from all three species of animals, confirming that this agent mediates anti-inflammatory effects in large animals. Although the results from this study were expected based on the precedence set in murine and human systems, it is important to understand the effects of administration of a compound or product in the species of interest as species-specific indications are not always available.


Asunto(s)
Antiinflamatorios/farmacología , Hidrocortisona/farmacología , Interferón gamma/biosíntesis , Leucocitos Mononucleares/efectos de los fármacos , Animales , Bovinos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Caballos , Leucocitos Mononucleares/inmunología , Ovinos , Especificidad de la Especie , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
5.
J Virol ; 76(5): 2141-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11836391

RESUMEN

Enzootic nasal tumor virus (ENTV) induces nasal epithelial cancer in infected sheep, but it is a simple retrovirus lacking a known oncogene. ENTV is closely related to jaagsiekte sheep retrovirus (JSRV), which also causes cancer in sheep but in the epithelial cells of the lower airways and alveoli. Here we show that as with JSRV, the envelope (Env) protein of ENTV can transform cultured cells and thus is likely to be responsible for oncogenesis in animals. In addition, the ENTV Env protein mediates virus entry using the same receptor as does JSRV Env, the candidate tumor suppressor Hyal2. However, ENTV Env mediates entry into cells from a more restricted range of species than does JSRV, and based on this finding we have identified amino acid regions in the Env proteins that are important for virus entry. Also, because ENTV does not efficiently use human Hyal2 as a receptor, we cloned the ovine Hyal2 cDNA and show that the encoded protein functions as an efficient receptor for both ENTV and JSRV. In summary, although ENTV and JSRV use the same cell surface receptor for cell entry and apparently transform cells by the same mechanism, they induce cancer in different tissues of infected sheep, indicating that oncogenesis is regulated at some other level. The transcriptional regulatory elements in these viruses are quite different, indicating that tissue-specific oncogenesis is likely regulated at the level of viral gene expression.


Asunto(s)
Transformación Celular Viral , Vectores Genéticos , Neoplasias Nasales/virología , Infecciones por Retroviridae/virología , Retroviridae/patogenicidad , Infecciones Tumorales por Virus/virología , Secuencia de Aminoácidos , Animales , Bovinos , Células Cultivadas , Perros , Fibroblastos/virología , Productos del Gen env/química , Productos del Gen env/genética , Productos del Gen env/metabolismo , Humanos , Retrovirus Ovino Jaagsiekte , Ratones , Datos de Secuencia Molecular , Adenomatosis Pulmonar Ovina/virología , Conejos , Ratas , Retroviridae/fisiología , Ovinos , Piel/citología , Transducción Genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA