Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cancer Res Clin Oncol ; 148(12): 3511-3520, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35962287

RESUMEN

PURPOSE: Prostate cancer can undergo curative effects by radical prostatectomy or radical radiotherapy. However, the best treatment for more aggressive high-risk prostate cancer remains controversial. Insufficient infiltration capacity and dysfunction are commonly occurrences in engineered T lymphocytes expressing chimeric antigen receptor (CAR-T), characterizing cancer immunotherapy failure. We conducted this study to investigate whether the combinative application of docetaxel and PSMA-CAR-T cells could be a more effective treatment to prostate cancer. METHODS: Expressions of prostate specific membrane antigen (PSMA) on prostate cancer cells were examined by Flow cytometry. The efficaciousness of PSMA-CAR-T was evaluated in vitro using ELISA and RTCA. The effect of intermixed therapy was assessed in vivo utilizing a human prostate cancer liver metastasis mouse model and a human prostate cancer cell xenograft mouse model. RESULTS: The outcome of cytokine discharge and cell killing assays demonstrated that PSMA-CAR-T cells have characteristic effector capacity against PSMA+ prostate cancer cells in vitro. Additionally, collaborative treatment of PSMA-CAR-T cells and docetaxel have cooperative efficacy in a mouse model of human prostate cancer. The merged strategy could be seen as an undeveloped avenue to augmenting adoptive CAR-T cell immunotherapy and mitigating the adverse side effects of chemotherapy. CONCLUSIONS: Cooperation of PSMA-specific CAR-T cells and the chemotherapy drug docetaxel can impressively ameliorate antitumor effectiveness against an installed metastatic human prostate cancer model in NPG mice.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias de la Próstata , Receptores Quiméricos de Antígenos , Masculino , Humanos , Ratones , Animales , Próstata/patología , Docetaxel , Células Supresoras de Origen Mieloide/patología , Neoplasias de la Próstata/tratamiento farmacológico , Inmunoterapia Adoptiva , Linfocitos T , Citocinas , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral
2.
Cell Immunol ; 372: 104475, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35063904

RESUMEN

M1 polarization of macrophages works as a promoter in pathogenesis of acute lung injury / acute respiratory distress syndrome (ALI/ARDS) by the secretion of pro-inflammatory cytokines and recruiting other inflammatory cells. Lipopolysaccharide (LPS), a critical component of the wall of gram-negative bacteria, can induce M1 polarization and ALI. Recently, cluster of differentiation 36 (CD36) has been reported to be associated with inflammatory responses. However, it has not yet been clarified whether CD36 in macrophages is involved in LPS-induced ALI. Herein, we demonstrated that in macrophages, LPS-induced ALI was regulated by CD36. Loss of CD36 attenuated LPS-induced ALI by reducing M1 polarization. Mechanistically, CD36 promoted macrophage M1 polarization by regulating CD14 associated with TLR4 during LPS stimulation. The findings of this study, clarified the mechanism of LPS-induced ALI through CD36 in macrophages, which provides a potential target for the prevention and treatment of ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Antígenos CD36/inmunología , Macrófagos Alveolares/clasificación , Macrófagos Alveolares/inmunología , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Traslado Adoptivo , Animales , Antígenos CD36/antagonistas & inhibidores , Antígenos CD36/genética , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos Alveolares/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Transducción de Señal , Receptor Toll-Like 4/metabolismo
3.
J Immunother ; 43(1): 16-28, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31574023

RESUMEN

Treatment with chimeric antigen receptor-modified T cell (CAR-T) has demonstrated promising therapeutic efficacy in hematologic malignancies. However, the therapeutic efficacy is still very limited for solid tumors. An immunosuppressive microenvironment is one of the main reasons for the limited efficacy. Some chemotherapeutic agents exhibit immune microenvironment modulation. Therefore, combination with chemotherapeutic agents may be one of the promising strategies to enhance the therapeutic efficacy of CAR-T against solid tumors. Sunitinib modulates the antitumor immune response by improving T-cell infiltration and function while reducing immunosuppressive factors. The authors constructed a second-generation CAR targeting human renal cell carcinoma (RCC)-specific antigen carbonic anhydrase IX (CAIX) with the costimulatory domain of 4-1BB. The results of cytokine releasing and cell killing assays showed that the CAIX-CAR-T cells have specific effector functions against CAIX renal cancer cells in vitro. Combination therapy with CAIX-CAR-T and sunitinib showed synergistic efficacy against a mouse lung metastasis model of human RCC. CAIX-CAR-T cells in the mice of the combination therapy group showed stronger proliferation and tumor infiltration than that in the mice of the CAIX-CAR-T monotherapy group. The possible mechanisms of the synergistic efficacy are: (1) sunitinib caused upregulation of CAIX in tumor cells; (2) sunitinib decreased frequency of myeloid-derived suppressor cells in the tumor microenvironment. Our study supplied an innovative immunotherapeutic approach whereby combining CAIX-CAR-T with sunitinib induces a potent antitumor response in an experimental model of metastatic RCC. The combination strategy should be considered as a potential approach to augment adoptive CAR-T cell immunotherapy.


Asunto(s)
Anhidrasa Carbónica IX/antagonistas & inhibidores , Inmunoterapia Adoptiva , Neoplasias Renales/patología , Neoplasias Renales/terapia , Sunitinib/farmacología , Linfocitos T/inmunología , Animales , Biomarcadores , Anhidrasa Carbónica IX/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Terapia Combinada , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Am J Cancer Res ; 9(7): 1382-1395, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31392076

RESUMEN

Based on previous reports, the efficacy of lenvatinib against cancer is mainly attributed to its antiangiogenic activity and its ability to suppress tumor proliferation, which are mediated by targeting receptor tyrosine kinases (RTKs). However, the effects of lenvatinib on tumor immune modulation have rarely been explored. Here, we show that lenvatinib effectively inhibited murine melanoma and renal cancer, and this inhibition was associated with enhanced tumor infiltration by natural killer (NK) cells. Critically, lenvatinib-induced tumor growth inhibition was attenuated by antibody-mediated NK cell depletion or the blockade of NK cell chemotaxis with an anti-CXCR3 blocking antibody. In addition, the expression of natural cytotoxicity receptors (NCRs) by tumor-infiltrating NK cells and the expression of cytotoxic cytokines in the tumor tissue were also augmented by lenvatinib. These data thus suggest that lenvatinib may be used not only as a direct cytotoxic drug against tumor angiogenesis and proliferation but also as a potent adjunct for enhancing the efficacy of immune-based cancer therapies by enhancing the tumor infiltration and activation of NK cells.

6.
J Immunol Res ; 2018: 4263520, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30410941

RESUMEN

Adoptive chimeric antigen receptor-modified T or NK cells (CAR-T or CAR-NK) offer new options for cancer treatment. CAR-T therapy has achieved encouraging breakthroughs in the treatment of hematological malignancies. However, their therapeutic efficacy against solid tumors is limited. New regimens, including combinations with chemical drugs, need to be studied to enhance the therapeutic efficacy of CAR-T or NK cells for solid tumors. An epithelial cell adhesion molecule- (EpCAM-) specific second-generation CAR was constructed and transduced into NK-92 cells by lentiviral vectors. Immune effects, including cytokine release and cytotoxicity of the CAR-NK-92 cells against EpCAM-positive colon cancer cells, were evaluated in vitro. Synergistic effects of regorafenib and CAR-NK-92 cells were analyzed in a mouse model with human colorectal cancer xenografts. The CAR-NK-92 cells can specifically recognize EpCAM-positive colorectal cancer cells and release cytokines, including IFN-γ, perforin, and granzyme B, and show specific cytotoxicity in vitro. The growth suppression efficacy of combination therapy with regorafenib and CAR-NK-92 cells on established EpCAM-positive tumor xenografts was more significant than that of monotherapy with CAR-NK-92 cells or regorafenib. Our results provided a novel strategy to treat colorectal cancer and enhance the therapeutic efficacy of CAR-modified immune effector cells for solid tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Vacunas contra el Cáncer/inmunología , Neoplasias Colorrectales/terapia , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/fisiología , Compuestos de Fenilurea/uso terapéutico , Piridinas/uso terapéutico , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Terapia Combinada , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Molécula de Adhesión Celular Epitelial/inmunología , Femenino , Humanos , Interferón gamma/metabolismo , Células Asesinas Naturales/trasplante , Ratones , Ratones SCID , Receptores Quiméricos de Antígenos/genética , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncol Rep ; 40(6): 3714-3724, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30272343

RESUMEN

Adoptive chimeric antigen receptor (CAR) T or NK cells offer new options for cancer treatment. Clinical results indicate that CAR­modified T cell (CAR­T) therapy has curative therapeutic efficacy for hematological malignancies. However, the efficacy of the therapy in most solid tumors, including advanced renal cell carcinoma (RCC), remains highly limited. New regimens, including combination of CAR­T cells with chemical drugs, must be studied to enhance the therapeutic efficacy of CAR­T or NK cells for solid tumors. In the present study, a carbonic anhydrase IX (CAIX)­specific third­generation CAR was transduced into NK92 cells by lentiviral vectors. The immune effects, including cytokine release and cytotoxicity, of the CAR­NK92 cells against CAIX­positive RCC cells were evaluated in vitro. Combination therapeutic effects of bortezomib and CAR­NK92 cells were analyzed in a mouse model with human RCC xenografts. The results revealed that CAIX­specific CAR­NK92 cells specifically recognized in vitro cultured CAIX­positive RCC cells and released cytokines, including IFN­Î³, perforin and granzyme B, and exhibited specific cytotoxicity. The cytotoxicity of the CAR­NK92 cells was enhanced after treating RCC cells with bortezomib in vitro. The suppressive efficacy of bortezomib combined with CAR­NK92 cells against established CAIX­positive tumor xenografts was more significant than that of the monotherapy with either CAR­NK92 cells or bortezomib. Therefore, bortezomib can enhance the effects of the CAR­NK92 cells against RCC in vitro and in vivo. This study provided an experimental basis for the novel clinical regimen of CAIX­specific CAR­modified NK or T cells for the treatment of RCC.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Bortezomib/administración & dosificación , Anhidrasa Carbónica IX/metabolismo , Carcinoma de Células Renales/terapia , Neoplasias Renales/terapia , Leucocitos Mononucleares/trasplante , Receptores Quiméricos de Antígenos/metabolismo , Animales , Bortezomib/farmacología , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Terapia Combinada , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia Adoptiva , Neoplasias Renales/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...