Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-37333207

RESUMO

Background: Osteosarcoma (OS) patients that present with metastatic disease have a poor prognosis and no curative options. Allogeneic bone marrow transplant (alloBMT) is curative for hematologic malignancies through the graft-versus-tumor (GVT) effect, but to date has been ineffective for solid tumors like OS. CD155 is expressed on OS and interacts strongly with the inhibitory receptors TIGIT and CD96 but also binds to the activating receptor DNAM-1 on natural killer (NK) cells but has never been targeted after alloBMT. Combining adoptive transfer of allogeneic NK (alloNK) cells with CD155 checkpoint blockade after alloBMT may enhance a GVT effect against OS but could enhance toxicities like graft-versus-host-disease (GVHD). Methods: Ex vivo activated and expanded murine NK cells were generated with soluble IL-15/IL- 15Rα. AlloNK and syngeneic NK (synNK) cell phenotype, cytotoxicity, cytokine production, and degranulation against the CD155-expressing murine OS cell line K7M2 were assessed in vitro. Mice bearing pulmonary OS metastases underwent alloBMT followed by infusion of alloNK cells with combinations of anti-CD155 and anti-DNAM-1 blockade. Tumor growth, GVHD and survival were monitored and differential gene expression of lung tissue was assessed by RNA microarray. Results: AlloNK cells exhibited superior cytotoxicity against CD155-expressing OS compared to synNK cells, and this activity was further enhanced by CD155 blockade. CD155 blockade increased alloNK cell degranulation and interferon gamma production through DNAM-1, as these functions were abrogated during DNAM-1 blockade. In vivo, CD155 blockade after alloBMT increased EFS with no exacerbation of GVHD. Treatment with combination CD155 and DNAM-1 blockade ameliorated survival and tumor control benefits seen with CD155 blockade alone. In mice treated with CD155 blockade, genes related to NK cell cytotoxicity were upregulated. DNAM-1 blockade resulted in upregulation of NK cell inhibition. Conclusions: These results demonstrate the safety and efficacy of infusing alloNK cells with CD155 blockade to mount a GVT effect against OS and show benefits are in part through DNAM-1. Defining the hierarchy of receptors that govern alloNK responses will be critical to translating combination adoptive NK cell and immune checkpoint inhibition for patients with solid tumors treated with alloBMT. WHAT IS ALREADY KNOWN ON THIS TOPIC: Allogeneic bone marrow transplant (alloBMT) has yet to show efficacy in treating solid tumors, such as osteosarcoma (OS). CD155 is expressed on OS and interacts with natural killer (NK) cell receptors, such as activating receptor DNAM-1 and inhibitory receptors TIGIT and CD96 and has a dominant inhibitory effect on NK cell activity. Targeting CD155 interactions on allogeneic NK cells could enhance anti-OS responses, but this has not been tested after alloBMT. WHAT THIS STUDY ADDS: CD155 blockade enhances allogeneic natural killer cell-mediated cytotoxicity against osteosarcoma and improved event-free survival after alloBMT in an in vivo mouse model of metastatic pulmonary OS. Addition of DNAM-1 blockade abrogated CD155 blockade-enhanced allogeneic NK cell antitumor responses. HOW THIS STUDY MIGHT AFFECT RESEARCH PRACTICE OR POLICY: These results demonstrate efficacy of allogeneic NK cells combined with CD155 blockade to mount an antitumor response against CD155-expressing OS. Translation of combination adoptive NK cell and CD155 axis modulation offers a platform for alloBMT treatment approaches for pediatric patients with relapsed and refractory solid tumors.

2.
Cancers (Basel) ; 13(11)2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-34199783

RESUMO

Treatment of metastatic pediatric solid tumors remain a significant challenge, particularly in relapsed and refractory settings. Standard treatment has included surgical resection, radiation, chemotherapy, and, in the case of neuroblastoma, immunotherapy. Despite such intensive therapy, cancer recurrence is common, and most tumors become refractory to prior therapy, leaving patients with few conventional treatment options. Natural killer (NK) cells are non-major histocompatibility complex (MHC)-restricted lymphocytes that boast several complex killing mechanisms but at an added advantage of not causing graft-versus-host disease, making use of allogeneic NK cells a potential therapeutic option. On top of their killing capacity, NK cells also produce several cytokines and growth factors that act as key regulators of the adaptive immune system, positioning themselves as ideal effector cells for stimulating heavily pretreated immune systems. Despite this promise, clinical efficacy of adoptive NK cell therapy to date has been inconsistent, prompting a detailed understanding of the biological pathways within NK cells that can be leveraged to develop "next generation" NK cell therapies. Here, we review advances in current approaches to optimizing the NK cell antitumor response including combination with other immunotherapies, cytokines, checkpoint inhibition, and engineering NK cells with chimeric antigen receptors (CARs) for the treatment of pediatric solid tumors.

3.
Leuk Lymphoma ; 62(5): 1167-1177, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33843403

RESUMO

Targeting the JAK/STAT and BCL2 pathways in patients with relapsed/refractory T cell acute lymphoblastic leukemia (T-ALL) may provide an alternative approach to achieve clinical remissions. Ruxolitinib and venetoclax show a dose-dependent effect on T-ALL individually, but combination treatment reduces survival and proliferation of T-ALL in vitro. Using a xenograft model, the combination treatment fails to improve survival, with death from hind limb paralysis. Despite on-target inhibition by the drugs, histopathology demonstrates increased leukemic infiltration into the central nervous system (CNS) as compared to liver or bone marrow. Liquid chromatography-tandem mass spectroscopy shows that ruxolitinib and venetoclax insufficiently cross into the CNS. The addition of the CXCR4 inhibitor plerixafor with ruxolitinib and venetoclax reduces clinical scores and enhances survival. While combination therapy with ruxolitinib and venetoclax shows promise for treating T-ALL, additional inhibition of the CXCR4-CXCL12 axis may be needed to maximize the possibility of complete remission.


Assuntos
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Receptores CXCR4 , Benzilaminas , Compostos Bicíclicos Heterocíclicos com Pontes , Sistema Nervoso Central , Ciclamos , Mobilização de Células-Tronco Hematopoéticas , Humanos , Janus Quinase 1 , Nitrilas , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/genética , Pirazóis , Pirimidinas , Sulfonamidas
4.
J Clin Invest ; 130(6): 2816-2819, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32391808

RESUMO

Programmed cell death protein 1 (PD-1) has become one of the most investigated targets for cancer immunotherapy. Most research has centered on inhibiting PD-1 on T cells, but there is increased interest in understanding the role of PD-1 on NK cells. While the expression of PD-1 on NK cells has been controversial, with papers publishing contradictory results in multiple models, there is increased clinical interest in NK and PD-1 immunotherapy. In this issue of the JCI, Judge et al. comprehensively explore the lack of PD-1 expression on murine, canine, and human NK cells and the clinical implication of these findings.


Assuntos
Antígeno B7-H1 , Receptor de Morte Celular Programada 1 , Animais , Cães , Humanos , Imunoterapia , Células Matadoras Naturais , Camundongos , Receptor de Morte Celular Programada 1/genética , Linfócitos T
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA