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.
MAbs ; 15(1): 2287250, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38047502

RESUMEN

PD-1 checkpoint inhibitors have revolutionized the treatment of patients with different cancer histologies including melanoma, renal cell carcinoma, and non-small cell lung carcinoma. However, only a subset of patients show a dramatic clinical response to treatment. Despite intense biomarker discovery efforts, no single robust, prognostic correlation has emerged as a valid outcome predictor. Immune competent, pet dogs develop spontaneous tumors that share similar features to human cancers including chromosome aberrations, molecular subtypes, immune signatures, tumor heterogeneity, metastatic behavior, and chemotherapeutic response. As such, they represent a valuable parallel patient population in which to investigate predictive biomarkers of checkpoint inhibition. However, the lack of a validated, non-immunogenic, canine anti-PD-1 antibody for pre-clinical use hinders this comparative approach and prevents potential clinical benefits of PD-1 blockade being realized in the veterinary clinic. To address this, fully canine single-chain variable fragments (scFvs) that bind canine (c)PD-1 were isolated from a comprehensive canine scFv phage display library. Lead candidates were identified that bound with high affinity to cPD-1 and inhibited its interaction with canine PD-L1 (cPD-L1). The lead scFv candidate re-formatted into a fully canine IgGD reversed the inhibitory effects of cPD-1:cPD-L1 interaction on canine chimeric antigen receptor (CAR) T cell function. In vivo administration showed no toxicity and revealed favorable pharmacokinetics for a reasonable dosing schedule. These results pave the way for clinical trials with anti-cPD-1 in canine cancer patients to investigate predictive biomarkers and combination regimens to inform human clinical trials and bring a promising checkpoint inhibitor into the veterinary armamentarium.


Asunto(s)
Melanoma , Investigación Biomédica Traslacional , Humanos , Perros , Animales , Receptor de Muerte Celular Programada 1 , Anticuerpos Monoclonales/uso terapéutico , Inhibidores de Puntos de Control Inmunológico , Antígeno B7-H1
2.
MAbs ; 13(1): 2004638, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34856888

RESUMEN

The immune checkpoint inhibitor (ICI) ipilimumab has revolutionized the treatment of patients with different cancer histologies, including melanoma, renal cell carcinoma, and non-small cell lung carcinoma. However, only a subset of patients shows dramatic clinical responses to treatment. Despite intense biomarker discovery efforts linked to clinical trials using CTLA4 checkpoint blockade, no single prognostic correlate has emerged as a valid predictor of outcome. Client-owned, immune competent, pet dogs develop spontaneous tumors that exhibit similar features to human cancers, including shared chromosome aberrations, molecular subtypes, immune signatures, tumor heterogeneity, metastatic behavior, and response to chemotherapy. As such, they represent a valuable parallel patient population in which to investigate novel predictive biomarkers and rational therapeutic ICI combinations. However, the lack of validated, non-immunogenic, canine ICIs for preclinical use hinders this comparative approach. To address this, fully canine single-chain variable fragments (scFvs) that bind canine CTLA4 were isolated from a comprehensive canine scFv phage display library. A lead candidate for clinical development was selected based on its subnanomolar binding affinity to canine CTLA4 and its ability to prevent CTLA4 binding to CD80/CD86 and promote T cell proliferation and effector function. In vivo mouse studies revealed pharmacokinetics similar to isotype control IgG with no evidence of short-term adverse effects. This work paves the way for in vivo analysis of the first fully canine, anti-canine CTLA4 antibody to promote anti-tumor immunity in dogs with immune-responsive cancers and provide an important comparative tool to investigate correlative biomarkers of response and mechanisms of resistance to CTLA4 checkpoint inhibition.


Asunto(s)
Neoplasias Pulmonares , Melanoma , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno CTLA-4 , Perros , Humanos , Ratones , Investigación Biomédica Traslacional
3.
Mol Cell Biol ; 27(5): 1947-59, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17210642

RESUMEN

Bloom's syndrome is a genetic disorder characterized by increased incidence of cancer and an immunodeficiency of unknown origin. The BLM gene mutated in Bloom's syndrome encodes a DNA helicase involved in the maintenance of genomic integrity. To explore the role of BLM in the immune system, we ablated murine Blm in the T-cell lineage. In the absence of Blm, thymocytes were severely reduced in numbers and displayed a developmental block at the beta-selection checkpoint that was partially p53 dependent. Blm-deficient thymocytes rearranged their T-cell receptor (TCR) beta genes normally yet failed to survive and proliferate in response to pre-TCR signaling. Furthermore, peripheral T cells were reduced in numbers, manifested defective homeostatic and TCR-induced proliferation, and produced extensive chromosomal damage. Finally, CD4(+) and CD8(+) T-cell responses were impaired upon antigen challenge. Thus, by ensuring genomic stability, Blm serves a vital role for development, maintenance, and function of T lymphocytes, suggesting a basis for the immune deficiency in Bloom's syndrome.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Linaje de la Célula , ADN Helicasas/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/metabolismo , Adenosina Trifosfatasas/genética , Alelos , Animales , Células Cultivadas , Aberraciones Cromosómicas , ADN Helicasas/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , RecQ Helicasas , Linfocitos T/inmunología , Timo/citología
4.
Mol Cell Biol ; 26(17): 6713-26, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16914751

RESUMEN

Bloom's syndrome (BS) is a genetic disorder characterized cellularly by increases in sister chromatid exchanges (SCEs) and numbers of micronuclei. BS is caused by mutation in the BLM DNA helicase gene and involves a greatly enhanced risk of developing the range of malignancies seen in the general population. With a mouse model for the disease, we set out to determine the relationship between genomic instability and neoplasia. We used a novel two-step analysis to investigate a panel of eight cell lines developed from mammary tumors that appeared in Blm conditional knockout mice. First, the panel of cell lines was examined for instability. High numbers of SCEs were uniformly seen in members of the panel, and several lines produced chromosomal instability (CIN) manifested by high numbers of chromosomal structural aberrations (CAs) and chromosome missegregation events. Second, to see if Blm mutation was responsible for the CIN, time-dependent analysis was conducted on a tumor line harboring a functional floxed Blm allele. The floxed allele was deleted in vitro, and mutant as well as control subclones were cultured for 100 passages. By passage 100, six of nine mutant subclones had acquired high CIN. Nine mutant subclones produced 50-fold more CAs than did nine control subclones. Finally, chromosome loss preceded the appearance of CIN, suggesting that this loss provides a potential mechanism for the induction of instability in mutant subclones. Such aneuploidy or CIN is a universal feature of neoplasia but has an uncertain function in oncogenesis. Our results show that Blm gene mutation produces this instability, strengthening a role for CIN in the development of human cancer.


Asunto(s)
Adenosina Trifosfatasas/genética , Inestabilidad Cromosómica/genética , ADN Helicasas/genética , Eliminación de Secuencia/genética , Alelos , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Células Cultivadas , Cromosomas de los Mamíferos/genética , Células Clonales , ADN/genética , Embrión de Mamíferos/citología , Exones/genética , Humanos , Integrasas/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales , Ratones , Ratones Noqueados , Células 3T3 NIH , Ploidias , RecQ Helicasas , Células Madre/metabolismo , Factores de Tiempo
5.
DNA Repair (Amst) ; 2(12): 1387-404, 2003 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-14642567

RESUMEN

Independent mouse models for Bloom syndrome (BS) exist, each thought to disrupt Blm gene function. However, animals bearing these alleles exhibit distinct phenotypes. Blm(tm1Ches) and Blm(tm1Grdn) homozygous mutant animals exhibit embryonic lethality while in another, Blm(tm3Brd), homozygosity yields viable, fertile animals with a cancer predisposition. Further characterization reveals the Blm(tm3Brd) allele to be a hypomorph, producing a diminished quantity of normal mRNA and protein. The Blm(tm3Brd) allele produces sufficient normal protein to rescue Blm(tm1Ches) lethality. Evaluation of viable animals reveals an inverse correlation between the quantity of Blm protein and the level of chromosome instability and a similar genotypic relationship for tumor predisposition indicating that Blm protein is rate limiting for maintaining genomic stability and the avoidance of tumors.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Síndrome de Bloom/genética , Inestabilidad Cromosómica , ADN Helicasas/metabolismo , Desarrollo Embrionario y Fetal/genética , Predisposición Genética a la Enfermedad , Neoplasias/genética , Animales , Pérdida del Embrión/genética , Femenino , Marcación de Gen , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , RecQ Helicasas , Bazo/metabolismo , Supresión Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA