Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 224
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cytotherapy ; 22(10): 552-562, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32747298

RESUMEN

BACKGROUND AIMS: Anti-CD19 chimeric antigen receptor (CAR)-modified T cells have shown dramatic cytotoxicity against B-cell malignancies. Currently, autologous T cells are conventionally used to manufacture CAR T cells. Low quality or insufficient quantity of autologous T cells may lead to failure of CAR T preparations. Moreover, CAR T preparation usually takes 1-2 weeks, which is too long for patients with rapid disease progression to successfully infuse CAR T cells. Thus, the development of a ready-to-use CAR immunotherapy strategy is needed. NK-92, a natural killer (NK) cell line derived from an NK lymphoma patient, has been gradually applied as a CAR-modified effector cell. To avoid the potential development of secondary NK lymphoma in patients, large doses of radiation are used to treat NK-92 cells before clinical application, which ensures the safety but reduces the cytotoxicity of NK-92 cells. Therefore, it is crucial to explore a suitable radiation dose that ensures short life span and good cytotoxicity of CAR NK-92 cells. METHODS: NK-92MI, a modified IL-2-independent NK-92 cell line, was used to establish an anti-CD19 CAR NK. The suitable radiation dose of CAR NK was then explored in vitro and validated in vivo, and the specific cytotoxicity of irradiated and unirradiated CAR NK against CD19+ malignant cells was assessed. RESULTS: CAR NK exhibited specific cytotoxicity against CD19+ malignant cells. Irradiation ensured a short life span of CAR NK in vitro and in vivo. Encouragingly, irradiated CAR NK displayed an anti-CD19+ malignancy capacity similar to that of unirradiated CAR NK. CONCLUSIONS: Five Gy is a suitable radiation dose to ensure the safety and effectiveness of CD19 CAR NK-92MI cells.


Asunto(s)
Antígenos CD19/metabolismo , Citotoxicidad Inmunológica , Receptores Quiméricos de Antígenos/metabolismo , Adulto , Anciano , Animales , Linfocitos B/inmunología , Linfocitos B/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular , Citotoxicidad Inmunológica/efectos de la radiación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de la radiación , Masculino , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Adulto Joven
2.
Cytotherapy ; 20(5): 715-727, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29572116

RESUMEN

BACKGROUND AIMS: Irradiation enhances the adhesion between natural killer (NK) cells and target cells by up-regulating intercellular adhesion molecule-1 (ICAM-1) on target cells. Therefore, we investigated the effect of irradiation-induced ICAM-1 expression on human cancer cells on NK cell-mediated cytotoxicity. METHODS: Expression levels of ICAM-1 on the target cell surface before and after irradiation of six human cancer cell lines (HL60, SKBR-3, T47D, HCT-116, U937 and U251) were analyzed by flow cytometry. Ex vivo expansion of NK cells from human peripheral blood mononuclear cells was performed by co-culture with irradiated K562 cells. The related adhesion molecule lymphocyte function-associated antigen 1 (LFA-1) on NK cells was analyzed by flow cytometry. An enzyme-linked immunosorbent assay was used to detect interferon-γ (IFN-γ), and WST-8 assays were performed to check NK cell cytotoxicity. Finally, blocking assays were performed using monoclonal antibodies against ICAM-1 or LFA-1. RESULTS: LFA-1 expression increased on NK cells after expansion (P <0.001). The expression of ICAM-1 was significantly upregulated by irradiation after 24 h in various cell lines, including HL60 (P <0.001), SKBR-3 (P <0.001), T47D (P <0.001) and U937 (P <0.001), although the level of expression depended on the cell line. ICAM-1 expression was extremely low before and after irradiation in U251 cells. NK cell-mediated cytotoxicity increased after irradiation of HL60 (P <0.001), SKBR-3 (P <0.001), T47D (P = 0.003), and U937 (P = 0.004) cells, in which ICAM-1 expression was significantly increased after irradiation. IFN-γ production by NK cells in response to HL60 (P <0.001) and T47D (P = 0.011) cells significantly increased after irradiation. NK cell-mediated cytotoxicity against irradiated SKBR-3 (P <0.001) and irradiated T47D cells (P = 0.035) significantly decreased after blocking of ICAM-1. Blocking of LFA-1 on NK cells resulted in reduced cytotoxicity against irradiated HL60 (P <0.001) and irradiated SKBR-3 (P <0.001). CONCLUSIONS: Irradiation upregulates ICAM-1 expression on the surface of human cancer cells and enhances activated NK cell-mediated cytotoxicity. Therefore, irradiation combined with NK cell therapy may improve the antitumor effects of NK cells.


Asunto(s)
Citotoxicidad Inmunológica/efectos de la radiación , Molécula 1 de Adhesión Intercelular/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/efectos de la radiación , Neoplasias/inmunología , Neoplasias/metabolismo , Radiación Ionizante , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Cinética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Regulación hacia Arriba/efectos de los fármacos
3.
Tumour Biol ; 39(3): 1010428317695961, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28349820

RESUMEN

Recurrence of breast cancer after radiotherapy may be partly explained by the presence of radioresistant cells. Thus, it would be desirable to develop an effective therapy against radioresistant cells. In this study, we demonstrated the intense antitumor activity of cytokine-induced killer cells against MCF-7 and radioresistant MCF-7 cells, as revealed by cytokine-induced killer-mediated cytotoxicity, tumor cell proliferation, and tumor invasion. Radioresistant MCF-7 cells were more susceptible to cytokine-induced killer cell killing. The stronger cytotoxicity of cytokine-induced killer cells against radioresistant MCF-7 cells was dependent on the expression of major histocompatibility complex class I polypeptide-related sequence A/B on radioresistant MCF-7 cells after exposure of cytokine-induced killer cells to sensitized targets. In addition, we demonstrated that cytokine-induced killer cell treatment sensitized breast cancer cells to chemotherapy via the downregulation of TK1, TYMS, and MDR1. These results indicate that cytokine-induced killer cell treatment in combination with radiotherapy and/or chemotherapy may induce synergistic antitumor activities and represent a novel strategy for breast cancer.


Asunto(s)
Neoplasias de la Mama/radioterapia , Tratamiento Basado en Trasplante de Células y Tejidos , Células Asesinas Inducidas por Citocinas/metabolismo , Recurrencia Local de Neoplasia/radioterapia , Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Células Asesinas Inducidas por Citocinas/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de la radiación , Femenino , Humanos , Células MCF-7 , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Tolerancia a Radiación , Timidina Quinasa/biosíntesis , Timidilato Sintasa/biosíntesis
4.
Cell Immunol ; 303: 16-23, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27113815

RESUMEN

Radiotherapy has been employed for the treatment of oncological patients for nearly a century, and together with surgery and chemotherapy, radiation oncology constitutes one of the three pillars of cancer therapy. Ionizing radiation has complex effects on neoplastic cells and on tumor microenvironment: beyond its action as a direct cytotoxic agent, tumor irradiation triggers a series of alterations in tumoral cells, which includes the de novo synthesis of particular proteins and the up/down-regulation of cell surface molecules. Additionally, ionizing radiation may induce the release of "danger signals" which may, in turn lead to cellular and molecular responses by the immune system. This immunomodulatory action of ionizing radiation highlights the importance of the combined use (radiotherapy plus immunotherapy) for cancer healing. Major histocompatibility complex antigens (also called Human Leukocyte Antigens, HLA in humans) are one of those molecules whose expression is modulated after irradiation. This review summarizes the modulatory properties of ionizing radiation on the expression of HLA class I (classical and non-classical) and class II molecules, with special emphasis in non-classical HLA-I molecules.


Asunto(s)
Antígenos HLA/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias/radioterapia , Radiación Ionizante , Linfocitos T Citotóxicos/inmunología , Citotoxicidad Inmunológica/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Antígenos HLA/genética , Antígenos HLA-G/genética , Antígenos HLA-G/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunidad/efectos de la radiación , Inmunomodulación , Neoplasias/genética , Neoplasias/metabolismo , Radioinmunoterapia , Microambiente Tumoral/efectos de la radiación , Antígenos HLA-E
5.
J Immunol ; 192(7): 3101-10, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24600032

RESUMEN

As a side effect of cancer radiotherapy, immune cells receive varying doses of radiation. Whereas high doses of radiation (>10 Gy) can lead to lymphopenia, lower radiation doses (2-4 Gy) represent a valid treatment option in some hematological cancers, triggering clinically relevant immunological changes. Based on our earlier observations, we hypothesized that lower radiation doses have a direct positive effect on T cells. In this study, we show that 0.6-2.4 Gy radiation enhances proliferation and IFN-γ production of PBMC or purified T cells induced by stimulation via the TCR. Radiation with 1.2 Gy also lowered T cell activation threshold and broadened the Th1 cytokine profile. Although radiation alone did not activate T cells, when followed by TCR stimulation, ERK1/2 and Akt phosphorylation increased above that induced by stimulation alone. These changes were followed by an early increase in glucose uptake. Naive (CD45RA(+)) or memory (CD45RA(-)) T cell responses to stimulation were boosted at similar rates by radiation. Whereas increased Ag-specific cytotoxic activity of a CD8(+) T cell line manifested in a 4-h assay (10-20% increase), highly significant (5- to 10-fold) differences in cytokine production were detected in 6-d Ag-stimulation assays of PBMC, probably as a net outcome of death of nonstimulated and enhanced response of Ag-stimulated T cells. T cells from patients receiving pelvic radiation (2.2-2.75 Gy) also displayed increased cytokine production when stimulated in vitro. We report in this study enhanced T cell function induced by synergistic radiation treatment, with potential physiological significance in a wide range of T cell responses.


Asunto(s)
Proliferación Celular/efectos de la radiación , Péptidos/inmunología , Linfocitos T/inmunología , Linfocitos T/efectos de la radiación , Secuencia de Aminoácidos , Línea Celular Tumoral , Células Cultivadas , Citotoxicidad Inmunológica/inmunología , Citotoxicidad Inmunológica/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Epítopos de Linfocito T/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Glucosa/inmunología , Glucosa/farmacocinética , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Antígenos Comunes de Leucocito/inmunología , Antígenos Comunes de Leucocito/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/efectos de la radiación , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Masculino , Fosforilación/inmunología , Fosforilación/efectos de la radiación , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/radioterapia , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
6.
Lancet Oncol ; 16(13): e498-509, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26433823

RESUMEN

Checkpoint blockade immunotherapy has received mainstream attention as a result of striking and durable clinical responses in some patients with metastatic disease and a reasonable response rate in many tumour types. The activity of checkpoint blockade immunotherapy is not restricted to melanoma or lung cancer, and additional indications are expected in the future, with responses already reported in renal cancer, bladder cancer, and Hodgkin's lymphoma among many others. Additionally, the interactions between radiation and the immune system have been investigated, with several studies describing the synergistic effects on local and distant tumour control when radiation therapy is combined with immunotherapy. Clinical enthusiasm for this approach is strengthened by the many ongoing trials combining immunotherapy with definitive and palliative radiation. Herein, we discuss the biological and mechanistic rationale behind combining radiation with checkpoint blockade immunotherapy, with a focus on the preclinical data supporting this potentially synergistic combination. We explore potential hypotheses and important considerations for clinical trial designs. Finally, we reintroduce the notion of radiosensitising immunotherapy, akin to radiosensitising chemotherapy, as a potential definitive therapeutic modality.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Quimioradioterapia/métodos , Inmunoterapia/métodos , Neoplasias/terapia , Tolerancia a Radiación , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de la radiación , Células Dendríticas/diagnóstico por imagen , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/patología , Dosis de Radiación , Radiografía , Resultado del Tratamiento
7.
J Immunol ; 190(4): 1873-81, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23315072

RESUMEN

Adoptive cell transfer (ACT) of ex vivo-activated autologous tumor-reactive T cells is currently one of the most promising approaches for cancer immunotherapy. Recent studies provided some evidence that IL-17-producing CD8(+) (Tc17) cells may exhibit potent antitumor activity, but the specific mechanisms have not been completely defined. In this study, we used a murine melanoma lung-metastasis model and tested the therapeutic effects of gp100-specific polarized type I CD8(+) cytotoxic T (Tc1) or Tc17 cells combined with autologous bone marrow transplantation after total body irradiation. Bone marrow transplantation combined with ACT of antitumor (gp100-specific) Tc17 cells significantly suppressed the growth of established melanoma, whereas Tc1 cells induced long-term tumor regression. After ACT, Tc1 cells maintained their phenotype to produce IFN-γ, but not IL-17. However, although Tc17 cells largely preserved their ability to produce IL-17, a subset secreted IFN-γ or both IFN-γ and IL-17, indicating the plasticity of Tc17 cells in vivo. Furthermore, after ACT, the Tc17 cells had a long-lived effector T cell phenotype (CD127(hi)/KLRG-1(low)) as compared with Tc1 cells. Mechanistically, Tc1 cells mediated antitumor immunity primarily through the direct effect of IFN-γ on tumor cells. In contrast, despite the fact that some Tc17 cells also secreted IFN-γ, Tc17-mediated antitumor immunity was independent of the direct effects of IFN-γ on the tumor. Nevertheless, IFN-γ played a critical role by creating a microenvironment that promoted Tc17-mediated antitumor activity. Taken together, these studies demonstrate that both Tc1 and Tc17 cells can mediate effective antitumor immunity through distinct effector mechanisms, but Tc1 cells are superior to Tc17 cells in mediating tumor regression.


Asunto(s)
Traslado Adoptivo/métodos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Interleucina-17/biosíntesis , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/métodos , Trasplante de Médula Ósea/patología , Linfocitos T CD8-positivos/efectos de la radiación , Citotoxicidad Inmunológica/efectos de la radiación , Interleucina-17/efectos de la radiación , Neoplasias Pulmonares/patología , Activación de Linfocitos/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Irradiación Corporal Total/métodos , Antígeno gp100 del Melanoma/biosíntesis , Antígeno gp100 del Melanoma/inmunología
8.
Int J Cancer ; 134(10): 2383-92, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24154990

RESUMEN

Radiation therapy (RT) continues to be a cornerstone in the treatment for many cancers. Unfortunately, not all individuals respond effectively to RT resulting clinically in two groups consisting of nonresponders (progressive disease) and responders (tumor control/cure). The mechanisms that govern the outcome of radiotherapy are poorly understood. Interestingly, a new paradigm has emerged demonstrating that the immune system mediates many of the antitumor effects of RT. Therefore, we hypothesized that the immune response following RT may dictate the efficacy of treatment. To examine this, we developed a tumor model that mirrors this clinically relevant phenomenon in which mice bearing Colon38, a colon adenocarcinoma, were treated locally with 15Gy RT resulting in both nonresponders and responders. More importantly, we were able to distinguish responders from nonresponders as early as 4 days post-RT allowing for the unique opportunity to identify critical events that ultimately determined the effectiveness of therapy. Intratumoral immune cells and interferon-gamma were increased in responsive tumors and licensed CD8 T cells to exhibit lytic activity against tumor cells, a response that was diminished in tumors refractory to RT. Combinatorial treatment with RT and the immunomodulatory cytokine IL-12 resulted in complete remission of cancer in 100% of cases compared to a cure rate of only 12% with RT alone. Similar data were obtained when IL-12 was delivered by microspheres. Therefore, the efficacy of RT may depend on the strength of the immune response induced after radiotherapy. Additionally, immunotherapy that further stimulates the immune cells may enhance the effectiveness of RT.


Asunto(s)
Adenocarcinoma/radioterapia , Linfocitos T CD8-positivos/efectos de la radiación , Neoplasias del Colon/radioterapia , Citotoxicidad Inmunológica/efectos de la radiación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Análisis de Varianza , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Quimioradioterapia , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/patología , Sistema Inmunológico/efectos de la radiación , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-12/farmacología , Ratones , Resultado del Tratamiento
9.
Cancer Immunol Immunother ; 63(3): 259-71, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24357146

RESUMEN

The need for an intact immune system for cancer radiation therapy to be effective suggests that radiation not only acts directly on the tumor but also indirectly, through the activation of host immune components. Recent studies demonstrated that endogenous type I interferons (type I IFNs) play a role in radiation-mediated anti-tumor immunity by enhancing the ability of dendritic cells to cross-prime CD8(+) T cells. However, it is still unclear to what extent endogenous type I IFNs contribute to the recruitment and function of CD8(+) T cells. Little is also known about the effects of type I IFNs on myeloid cells. In the current study, we demonstrate that type I and type II IFNs (IFN-γ) are both required for the increased production of CXCL10 (IP-10) chemokine by myeloid cells within the tumor after radiation treatment. Radiation-induced intratumoral IP-10 levels in turn correlate with tumor-infiltrating CD8(+) T cell numbers. Moreover, type I IFNs promote potent tumor-reactive CD8(+) T cells by directly affecting the phenotype, effector molecule production, and enhancing cytolytic activity. Using a unique inducible expression system to increase local levels of IFN-α exogenously, we show here that the capacity of radiation therapy to result in tumor control can be enhanced. Our preclinical approach to study the effects of local increase in IFN-α levels can be used to further optimize the combination therapy strategy in terms of dosing and scheduling, which may lead to better clinical outcome.


Asunto(s)
Linfocitos T CD8-positivos/efectos de la radiación , Interferón-alfa/metabolismo , Neoplasias Mamarias Animales/radioterapia , Melanoma Experimental/inmunología , Melanoma Experimental/radioterapia , Células Mieloides/efectos de los fármacos , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/efectos de la radiación , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/efectos de la radiación , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Interferón-alfa/genética , Interferón-alfa/farmacología , Interferón gamma/genética , Interferón gamma/metabolismo , Recuento de Linfocitos , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Trasplante de Neoplasias
10.
Am J Pathol ; 182(6): 2345-54, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583648

RESUMEN

Cancer treatments using ionizing radiation (IR) therapy are thought to act primarily through the induction of tumor cell damage at a molecular level. However, a new concept has recently emerged, suggesting that the immune system is required for effective IR therapy. Our work here has identified interferon gamma (IFN-γ) as an essential cytokine for the efficacy of IR therapy. Local IR (15 Gy) to mice bearing Colon38, a colon adenocarcinoma, decreases tumor burden in wild-type animals. Interestingly, IR therapy had no effect on tumor burden in IFNγKO mice. We further determined that intratumoral levels of IFN-γ increased 2 days following IR, which directly correlated with a decrease in tumor burden that was not a result of direct cytotoxic effects of IFN-γ on tumor cells. T cells from IR-treated tumors exhibited a far greater capacity to lyse tumor cells in a (51)Cr release assay, a process that was dependent on IFN-γ. CD8(+) T cells were the predominant producers of IFN-γ, as demonstrated by IFN-γ intracellular staining and studies in IFN-γ reporter mice. Elimination of CD8(+) T cells by antibody treatment reduced the intratumoral levels of IFN-γ by over 90%. More importantly, elimination of CD8(+) T cells completely abrogated the effects of radiation therapy. Our data suggest that IFN-γ plays a pivotal role in mediating the antitumor effects of IR therapy.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/radioterapia , Neoplasias del Colon/inmunología , Neoplasias del Colon/radioterapia , Interferón gamma/inmunología , Adenocarcinoma/patología , Animales , Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/patología , Citotoxicidad Inmunológica/efectos de la radiación , Interferón gamma/biosíntesis , Interferón gamma/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias
11.
Tsitologiia ; 55(7): 501-6, 2013.
Artículo en Ruso | MEDLINE | ID: mdl-25509119

RESUMEN

Tumorigenicity of murine hepatoma cells (MH22a) and their sensitivity to lysis by natural killers (NKs) have been studied after exposure to polychromatic visible and infrared light (VIS-IR, 480-3400 nm, 40 mW/cm2), similar to the terrestrial solar spectrum without its minor UV component, in order to elucidate the involvement of this important environmental and physiotherapeutic factor in regulation of the anti-tumor defense system. The MH22 cells were in vitro exposed to VIS-IR light and their sensitivity to lytic activity of NKs was evaluated. We found that sensitivity of MH22a cells to lysis by NKs after exposure to VIS-IR light at a dose of 4.8 J/cm2 increased 1.5-2 times, while it did not change after exposure to a dose of 9.6 J/cm2 at all ratios (1 : 5-1 : 50) of the number of NKs (effectors) to that of hepatoma cells (targets). The increase in the sensitivity of hepatoma cells to NKs was accompanied by structural changes of cell surface: the capability of supramembraneous glycoproteins (glycocalix) to sorb the vital dye alcian blue (AB) was significantly lower as compared with the unexposed cells of control group. However, no changes in AB sorption was revealed in hepatoma cells exposed to the light at a dose 9.6 J/cm2. Tumorigenicity of photo-irradiated MH22a cells has been studied in the in vivo experiments. Light-exposed (4.8 and 9.6 J/cm2) and intact hepatoma cells were transplanted into syngenic mice C3HA. Tumor volumes 25 days after transplantation proved to be smaller after exposure to the light at both doses than in the control group (4-4.5 times and 2.5-4 times, respectively), which correlated with the increase in the sensitivity to lisys by NKs and decrease in the AB sorption only after light exposure at dose 4.8 J/cm2. Using the flow cytometry method we could show that VIS-IR light at the applied doses did not interfere with the distribution of hepatoma cells over the cycle phases and thus deceleration of the tumor growth was not associated with cytostatic effect of VIS-IR light. To evaluate effect of polychromatic light on the growth of the preformed tumors, the 5-day course of daily light exposures of tumor bearing mice C3HA was carried out in 10 days after subcutaneous transplantation of 2 x 10(5) cells of syngene hepatoma when the tumors had developed in 100% animals. Like in the case of transplantation of the light-exposed cells, irradiation of the tumor bearing mice at doses 4.8-9.6 J/cm2 resulted in deceleration of tumor growth (2.1-2.9 and 2.2 times respectively) for 4 weeks as compared with non-irradiated mice.


Asunto(s)
Carcinoma Hepatocelular/radioterapia , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/radioterapia , Células Tumorales Cultivadas/efectos de la radiación , Azul Alcián/metabolismo , Animales , Pruebas de Carcinogenicidad , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Citotoxicidad Inmunológica/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Glicocálix/química , Glicocálix/efectos de la radiación , Rayos Infrarrojos , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de la radiación , Luz , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C3H , Trasplante de Neoplasias , Carga Tumoral , Células Tumorales Cultivadas/trasplante
12.
J Exp Med ; 155(6): 1858-63, 1982 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-6978924

RESUMEN

Low dose radiosensitivity of in vitro generated alloimmune murine cytotoxic T lymphocytes (CTL) was studied. It appears that a subset of CTL exists that can be killed with 10-25 rad of x rays. These radiosensitive CTL are Lyt-1,2+ T lymphocytes. Analyses of cytotoxicity by chromium release assays indicate that the radiosensitive CTL are present in responder spleen cell cultures from all strains of mice tested. The generation of these effector cells is most pronounced in animals of the C57BL background. The mechanism of low dose radiosensitivity appears to be interphase death.


Asunto(s)
Citotoxicidad Inmunológica/efectos de la radiación , Linfocitos T/efectos de la radiación , Animales , Antígenos Ly/inmunología , Antígenos Ly/efectos de la radiación , Bromodesoxiuridina/metabolismo , Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Inmunidad Celular/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Fenotipo , Especificidad de la Especie , Linfocitos T/citología , Linfocitos T/inmunología
13.
J Exp Med ; 164(1): 180-95, 1986 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-3014033

RESUMEN

The role of HLA-DR+ cells in NK activity against CMV-infected FS4 foreskin fibroblasts and K562 erythroleukemia cells was examined. When nonadherent PBMC were depleted of either HLA-DR+ or Leu-11b+ cells by treatment with mAbs plus C, NK activity against CMV-FS4 target cells was markedly reduced. In contrast, depletion of HLA-DR+ cells had no effect on NK activity against K562 target cells. When HLA-DR-depleted cells were added to Leu-11b-depleted cells, NK activity against CMV-FS4 was restored. Negative selection experiments indicated that the HLA-DR+ cells contributing to NK activity against CMV-FS4 are not B or T cells, while negative and positive selection experiments excluded a role for monocytes. Experiments in which HLA-DR- and Leu-11b- cells were mixed in varying proportions indicated that NK(CMV-FS4) is mediated by Leu-11b+ cells, while HLA-DR+ cells provide an accessory function. Irradiation (50 GY) abolished the NK effector function of Leu-11b+ cells, but not the accessory function of HLA-DR+ cells. The NK activity against CMV-FS4 of HLA-DR- cells was restored by the addition of rIFN-alpha or of cell-free supernatants generated by coculturing PBMC or Leu-11b- cells with CMV-FS4. The ability of these supernatants to restore NK activity of HLA-DR- cells was completely abrogated by the addition of neutralizing amounts of antibody to IFN-alpha. In related experiments, neutralization of IFN-alpha in NK assays had little or no effect on NK activity against CMV-FS4, suggesting that the accessory function of HLA-DR+ cells might be mediated by alternative mechanisms in addition to the secretion of extracellular IFN-alpha.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Citomegalovirus/inmunología , Citotoxicidad Inmunológica , Fibroblastos/inmunología , Antígenos de Histocompatibilidad Clase II , Células Asesinas Naturales/inmunología , Células Presentadoras de Antígenos/clasificación , Células Presentadoras de Antígenos/efectos de la radiación , Antígenos de Diferenciación de Linfocitos T , Antígenos de Superficie , Sistema Libre de Células , Pruebas Inmunológicas de Citotoxicidad , Citotoxicidad Inmunológica/efectos de la radiación , Antígenos HLA-DR , Humanos , Sueros Inmunes/farmacología , Interferón Tipo I/inmunología , Interferón Tipo I/fisiología , Fenotipo
14.
Sci Adv ; 6(13): eaay9789, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32232155

RESUMEN

Radiotherapy (RT) is routinely used in cancer treatment, but expansion of its clinical indications remains challenging. The mechanism underlying the radiation-induced bystander effect (RIBE) is not understood and not therapeutically exploited. We suggest that the RIBE is predominantly mediated by irradiated tumor cell-released microparticles (RT-MPs), which induce broad antitumor effects and cause immunogenic death mainly through ferroptosis. Using a mouse model of malignant pleural effusion (MPE), we demonstrated that RT-MPs polarized microenvironmental M2 tumor-associated macrophages (M2-TAMs) to M1-TAMs and modulated antitumor interactions between TAMs and tumor cells. Following internalization of RT-MPs, TAMs displayed increased programmed cell death ligand 1 (PD-L1) expression, enhancing follow-up combined anti-PD-1 therapy that confers an ablative effect against MPE and cisplatin-resistant MPE mouse models. Immunological memory effects were induced.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Reprogramación Celular/inmunología , Citotoxicidad Inmunológica , Neoplasias/inmunología , Neoplasias/metabolismo , Radiación Ionizante , Animales , Biomarcadores , Biomarcadores de Tumor , Efecto Espectador/inmunología , Efecto Espectador/efectos de la radiación , Línea Celular Tumoral , Reprogramación Celular/efectos de la radiación , Citotoxicidad Inmunológica/efectos de la radiación , Modelos Animales de Enfermedad , Humanos , Memoria Inmunológica , Quinasas Janus/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Neoplasias/patología , Neoplasias/terapia , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Epigenomics ; 12(5): 397-408, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32267172

RESUMEN

Aim: We here hypothesized that tumor-derived exosomal miRNA (TexomiR) released from irradiated tumors may play a role in the tumor cells escape to natural killer (NK) cells. Materials & methods: Our study included the use of different cancer cell lines, blood biopsies of xenograph mice model and patients treated with radiotherapy. Results: The irradiation of cancer cells promotes the TET2-mediated demethylation of miR-378 promoter, miR-378a-3p overexpression and its loading in exosomes, inducing the decrease of granzyme-B (GZMB) secretion by NK cells. An inverse correlation between TexomiR-378a-3p and GZMB was observed in murine and human blood samples. Conclusion: Our work identifies TexomiR-378a-3p as a molecular signature associated with the loss of NK cells cytotoxicity via the decrease of GZMB expression upon radiotherapy.


Asunto(s)
Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , MicroARNs/genética , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/efectos de la radiación , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Expresión Génica , Granzimas/metabolismo , Humanos , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Radioterapia
16.
Sci Adv ; 6(8): eaay9209, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32128416

RESUMEN

T cells engineered to express chimeric antigen receptors (CARs) can recognize and engage with target cancer cells with redirected specificity for cancer immunotherapy. However, there is a lack of ideal CARs for solid tumor antigens, which may lead to severe adverse effects. Here, we developed a light-inducible nuclear translocation and dimerization (LINTAD) system for gene regulation to control CAR T activation. We first demonstrated light-controllable gene expression and functional modulation in human embryonic kidney 293T and Jurkat T cell lines. We then improved the LINTAD system to achieve optimal efficiency in primary human T cells. The results showed that pulsed light stimulations can activate LINTAD CAR T cells with strong cytotoxicity against target cancer cells, both in vitro and in vivo. Therefore, our LINTAD system can serve as an efficient tool to noninvasively control gene activation and activate inducible CAR T cells for precision cancer immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva , Luz , Neoplasias/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/efectos de la radiación , Animales , Antígenos CD19/inmunología , Antígenos de Neoplasias/inmunología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Criptocromos/genética , Criptocromos/metabolismo , Citotoxicidad Inmunológica/inmunología , Citotoxicidad Inmunológica/efectos de la radiación , Humanos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Ratones , Modelos Biológicos , Unión Proteica , Multimerización de Proteína , Receptores Quiméricos de Antígenos/genética , Activación Transcripcional/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Immunopathol Pharmacol ; 22(4): 1059-66, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20074470

RESUMEN

In a preliminary study a reduction in natural killer (NK) cell activity in peripheral blood lymphocytes (PBL) was observed in a group of workers exposed to levels of extremely low frequency-magnetic fields (ELF-MF) exceeding 1 microT. This study was performed to confirm the results. In 121 workers engaged in various occupational activities, individual ELF-MF exposure was monitored for 2 work shifts. Exposure levels were calculated as time-weighted average (TWA). Subjects were classified as Low exposure (TWA < or = 0.2 microT), Medium exposure (TWA 0.21-0.99 microT), or Higher exposure (TWA > or = 1 microT). In higher exposure workers NK activity proved significantly reduced compared to low exposure,(p<0.01). In medium exposure a reduction was also observed, but the difference was not significant. Multivariate analysis also confirmed the relation between exposure and NK activity. It has been suggested that ELF might affect tumour progression by inducing changes in the immune system: due to the role played by NK activity in host defence against cancer, the interference with the NK cell activity observed in this study is in agreement with this hypothesis. Furthermore, an increased risk for some neurodegenerative disorders has been reported in some epidemiological studies in ELF-MF-exposed workers: changes in NK function were also described in these diseases. Our results, showing the effect on NK activity of exposure exceeding 1 microT, suggest a possible mechanism for ELF-MF effects. This could open new horizons regarding the adverse long-term effects of these fields.


Asunto(s)
Citotoxicidad Inmunológica/efectos de la radiación , Campos Electromagnéticos/efectos adversos , Células Asesinas Naturales/efectos de la radiación , Exposición Profesional , Salud Laboral , Adulto , Técnicas de Cocultivo , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Medición de Riesgo , Factores de Tiempo
18.
J Clin Invest ; 129(10): 4224-4238, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31483286

RESUMEN

Treatment of tumors with ionizing radiation stimulates an antitumor immune response partly dependent on induction of IFNs. These IFNs directly enhance dendritic cell and CD8+ T cell activity. Here we show that resistance to an effective antitumor immune response is also a result of IFN signaling in a different cellular compartment of the tumor, the cancer cells themselves. We abolished type I IFN signaling in cancer cells by genetic elimination of its receptor, IFNAR1. Pronounced immune responses were provoked after ionizing radiation of tumors from 4 mouse cancer cell lines with Ifnar1 knockout. This enhanced response depended on CD8+ T cells and was mediated by enhanced susceptibility to T cell-mediated killing. Induction of Serpinb9 proved to be the mechanism underlying control of susceptibility to T cell killing after radiation. Ifnar1-deficient tumors had an augmented response to anti-PD-L1 immunotherapy with or without radiation. We conclude that type I IFN can protect cancer cells from T cell-mediated cytotoxicity through regulation of Serpinb9. This result helps explain why radiation of tumors can stimulate antitumor immunity yet also result in resistance. It further suggests potential targets for intervention to improve therapy and to predict responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Interferón Tipo I/inmunología , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/radioterapia , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/radioterapia , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/radioterapia , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Serpinas/genética , Serpinas/inmunología , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
19.
Radiats Biol Radioecol ; 48(6): 741-7, 2008.
Artículo en Ruso | MEDLINE | ID: mdl-19178053

RESUMEN

The influence of UV-light (240-390 nm) in dose 75.5-1510 J/m2 on the structural functional qualities (cytotoxic activity, antibody-produced ability, a level of expression of superficial receptors, a level cAMP) of lymphocytes peripheral blood of donors was investigated. Corregative action of UV-radiation on cytotoxic activity of lymphocytes in relation to cells of Ehrlich ascite carcinoma was revealed. The interrelation of modification types of plasmatic membrane components with a level of functional activity of lymphocytes was established. Was shown, that UV-light induces the decrease in a secondary level of messenger cAMP in cytosol of the photomodified cells. It is revealed, that UV-radiation causes stimulation of antibody-independent mechanism of cytotoxic lymphocytes, which is connected with activation of T-cells and with inhibition of antibody-dependent mechanism of cytotoxic activity.


Asunto(s)
Linfocitos B/efectos de la radiación , Citotoxicidad Inmunológica/efectos de la radiación , Linfocitos T/efectos de la radiación , Rayos Ultravioleta , Animales , Formación de Anticuerpos , Antígenos de Superficie/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Carcinoma de Ehrlich/inmunología , AMP Cíclico/metabolismo , Relación Dosis-Respuesta en la Radiación , Humanos , Activación de Linfocitos , Tolerancia a Radiación , Linfocitos T/inmunología , Linfocitos T/metabolismo
20.
J Clin Invest ; 71(6): 1737-43, 1983 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-6223049

RESUMEN

Recent studies of cartilage-hair hypoplasia (CHH), a form of short-limbed dwarfism, have shown that all affected individuals have a cellular proliferation defect that results in a cellular immunodeficiency. However, only a minority of CHH individuals suffer from severe, life-threatening infections. For this reason, relevant immune defense mechanisms that may be responsible for maintaining intact host defenses in the majority of CHH individuals were studied. Spontaneous and allogeneic culture-induced (mixed lymphocyte response-MLR) specific and nonspecific (NK-like) cytotoxic mechanisms were analyzed and correlated with lymphocyte subpopulations present in CHH and normal individuals. Spontaneous natural-killer (NK) activity was present at or above normal levels, but culture-induced specific cytotoxicity and NK-like cytotoxicity as well as NK-like activity by T cell lines were significantly reduced in CHH individuals. The generation of radiation-resistant cytotoxicity, which normally occurs during allogeneic MLR, was markedly diminished in CHH, and was correlated with the decreased proliferation observed in CHH cultures. Preservation of spontaneous NK activity and loss of all forms of culture-induced cytotoxicity was associated with an increase in the proportion of lymphocytes bearing a thymic independent NK phenotype (OKM1+ OKT3- Fc gamma + low-affinity E+), and a significant decrease in thymic derived OKT3+ cytolytic T cell sub-populations in CHH individuals. Therefore, an intact cellular cytotoxic effector mechanism has been identified in CHH (i.e., NK activity). Natural cytotoxicity may be of importance in maintaining host resistance to viral infections despite diminished thymic-derived effector mechanisms in cartilage-hair hypoplasia.


Asunto(s)
Citotoxicidad Inmunológica , Enanismo/inmunología , Células Asesinas Naturales/inmunología , Adolescente , Adulto , Anticuerpos Monoclonales , Antígenos de Superficie , Línea Celular , Citotoxicidad Inmunológica/efectos de la radiación , Enanismo/genética , Etnicidad , Humanos , Recuento de Leucocitos , Prueba de Cultivo Mixto de Linfocitos , Fenotipo , Receptores Fc/análisis , Linfocitos T/citología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA