Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Nat Methods ; 18(11): 1294-1303, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34725485

RESUMEN

Spheroids are three-dimensional cellular models with widespread basic and translational application across academia and industry. However, methodological transparency and guidelines for spheroid research have not yet been established. The MISpheroID Consortium developed a crowdsourcing knowledgebase that assembles the experimental parameters of 3,058 published spheroid-related experiments. Interrogation of this knowledgebase identified heterogeneity in the methodological setup of spheroids. Empirical evaluation and interlaboratory validation of selected variations in spheroid methodology revealed diverse impacts on spheroid metrics. To facilitate interpretation, stimulate transparency and increase awareness, the Consortium defines the MISpheroID string, a minimum set of experimental parameters required to report spheroid research. Thus, MISpheroID combines a valuable resource and a tool for three-dimensional cellular models to mine experimental parameters and to improve reproducibility.


Asunto(s)
Biomarcadores de Tumor/genética , Proliferación Celular , Bases del Conocimiento , Neoplasias/patología , Programas Informáticos , Esferoides Celulares/patología , Microambiente Tumoral , Técnicas de Cultivo de Célula/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/clasificación , Neoplasias/metabolismo , RNA-Seq , Reproducibilidad de los Resultados , Esferoides Celulares/inmunología , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas
2.
J Cell Mol Med ; 25(7): 3312-3326, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33634564

RESUMEN

Cancer stem cells (CSCs) are responsible for therapeutic resistance and recurrence in colorectal cancer. Despite advances in immunotherapy, the inability to specifically eradicate CSCs has led to treatment failure. Hence, identification of appropriate antigen sources is a major challenge in designing dendritic cell (DC)-based therapeutic strategies against CSCs. Here, in an in vitro model using the HT-29 colon cancer cell line, we explored the efficacy of DCs loaded with exosomes derived from CSC-enriched colonospheres (CSCenr -EXOs) as an antigen source in activating CSC-specific T-cell responses. HT-29 lysate, HT-29-EXOs and CSCenr lysate were independently assessed as separate antigen sources. Having confirmed CSCs enrichment in spheroids, CSCenr -EXOs were purified and characterized, and their impact on DC maturation was investigated. Finally, the impact of the antigen-pulsed DCs on the proliferation rate and also spheroid destructive capacity of autologous T cells was assessed. CSCenr -EXOs similar to other antigen groups had no suppressive/negative impacts on phenotypic maturation of DCs as judged by the expression level of costimulatory molecules. Notably, similar to CSCenr lysate, CSCenr -EXOs significantly increased the IL-12/IL-10 ratio in supernatants of mature DCs. CSCenr -EXO-loaded DCs effectively promoted T-cell proliferation. Importantly, T cells stimulated with CSCenr -EXOs disrupted spheroids' structure. Thus, CSCenr -EXOs present a novel and promising antigen source that in combination with conventional tumour bulk-derived antigens should be further explored in pre-clinical immunotherapeutic settings for the efficacy in hampering recurrence and metastatic spread.


Asunto(s)
Células Dendríticas/inmunología , Exosomas/inmunología , Inmunoterapia/métodos , Células Madre Neoplásicas/inmunología , Esferoides Celulares/inmunología , Células Cultivadas , Células HT29 , Humanos , Interleucinas/metabolismo , Esferoides Celulares/citología , Linfocitos T/inmunología
3.
Biochem Biophys Res Commun ; 552: 164-169, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33751933

RESUMEN

Human mesenchymal stem cells (MSCs) are known to have anti-inflammatory and immunomodulatory functions; thus, several MSC products have been applied as cell therapy in clinical trials worldwide. Recent studies have demonstrated that MSC spheroids have superior anti-inflammatory and immunomodulatory functions to a single cell suspension. Current methods to prepare MSC spheroids include hanging drop, concave microwell aggregation, spinner flask, and gravity circulation. However, all these methods have limitations such as low scalability, easy cell clumping, low viability, and irregular size distribution. Here, we present a nano-patterned culture plasticware named PAMcell™ 3D plate to overcome these limitations. Nano-sized silica particles (700 nm) coated with RGD peptide were arrayed into fusiform onto the PLGA film. This uniform array enabled the seeded MSCs to grow only on the silica particles, forming uniform-sized semi-spheroids within 48 h. These MSC spheroids have been shown to have enhanced stemness, anti-inflammatory, and immunomodulatory functions, as revealed by the increased expression of stem cell markers (Oct4, Sox2, and Nanog), anti-inflammatory (IL-10, TSG6, and IDO), and immunomodulatory molecules (HGF, VEGF, CXCR4) both at mRNA and protein expression levels. Furthermore, these MSC spheroids demonstrated an increased palliative effect on glycemic control in a multiple low-dose streptozotocin-induced diabetes model compared with the same number of MSC single cell suspensions. Taken together, this study presents a new method to produce uniform-sized MSC spheroids with enhanced anti-inflammatory and immunomodulatory functions in vitro and in vivo.


Asunto(s)
Tejido Adiposo/citología , Antiinflamatorios/inmunología , Técnicas de Cultivo de Célula/métodos , Factores Inmunológicos/inmunología , Células Madre Mesenquimatosas/inmunología , Esferoides Celulares/inmunología , Animales , Técnicas de Cultivo de Célula/instrumentación , Células Cultivadas , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/terapia , Expresión Génica/inmunología , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Esferoides Celulares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
4.
Mol Pharm ; 18(8): 2935-2946, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34191525

RESUMEN

The adoptive transfer of natural killer (NK) cells, which can recognize and obliterate cancer cells, provides a practical alternative to current treatment modalities to improve cancer patients' survival. However, translating NK cell therapies to treat solid tumors has proven challenging due to the tumor microenvironment (TME). Hypoxia in the TME induces immunosuppression that inhibits the cytotoxic function of NK cells. Thus, reversing hypoxia-induced immunosuppression is critical for effective adoptive NK cell immunotherapy. In this study, we use manganese dioxide nanoparticles (MnO2 NPs) to catalyze the degradation of tumor-produced hydrogen peroxide, thereby generating oxygen. For improved biocompatibility and modulation of oxygen production, the MnO2 NPs were encapsulated into poly(lactic-co-glycolic) to produce particles that are 116 nm in size and with a ζ-potential of +17 mV (PLGA-MnO2 NPs). The PLGA-MnO2 NPs showed first-order oxygen production and sustained high oxygen tension compared to equivalent amounts of bare MnO2 NPs in the presence of H2O2. The PLGA-MnO2 NPs were biocompatible, reduced hypoxia after penetration into the core of cancer spheroids, and decreased hypoxia-induced factor 1 α expression. Reducing hypoxia in the spheroid resulted in a decrease in the potent immunosuppressors, adenosine, and lactate, which was confirmed by electrospray ionization mass spectroscopy (ESI-MS). ESI-MS also showed a change in the metabolism of the amino acids aspartate, glutamine, and glutamate after hypoxia reduction in the cancer cells. Notably, the spheroids' microenvironment changes enhanced NK cells' cytotoxicity, which obliterated the spheroids. These results demonstrate that reducing hypoxia-induced immunosuppression in tumors is a potent strategy to increase the potency of cytotoxic immune cells in the TME. The developed NPs are promising new tools to improve adoptive NK cell therapy.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Células Asesinas Naturales/inmunología , Compuestos de Manganeso/farmacología , Nanopartículas del Metal/química , Nanocápsulas/química , Óxidos/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Esferoides Celulares/inmunología , Adenosina/metabolismo , Traslado Adoptivo/métodos , Neoplasias de la Mama/patología , Hipoxia de la Célula/inmunología , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Ácido Láctico/metabolismo , Células MCF-7 , Compuestos de Manganeso/química , Óxidos/química , Tamaño de la Partícula , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
5.
Exp Cell Res ; 390(2): 111968, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32197932

RESUMEN

EBV-associated gastric carcinoma (EBVaGC) is accompanied by massive lymphocyte infiltration, but therapy resistance and tumor progression still occur in patients with EBVaGC. Cancer stem cells (CSCs) are reported to possess immunomodulatory ability that allows them to resist immune-mediated rejection for many tumor types. However, whether and how CSCs in EBVaGC exhibit immunosuppression has not yet been elucidated. We isolated CSC-like sphere-forming cells (SFCs) from EBVaGC cell line SNU-719 using the cancer sphere method. We validated their CSC-associated properties in the expression of the epithelial-mesenchymal transition (EMT)-related genes, the ability to form colonies, and resistance to chemotherapy drug-induced apoptosis and explored their immunomodulatory ability using the coculture system with PBMC (peripheral blood mononuclear cell). These CSC-like SFCs were CD44+CD24-/low and were more tumorigenic than the parental SNU-719 cells in the xenograft mouse model. Remarkably, in the tumor-PBMC co-culturing experiments, these EBVaGC SFCs demonstrated profound immunosuppression by inhibiting the proliferation of PBMCs and T cell activation as well as inducing the generation of regulatory T cells (Tregs). Furthermore, the induction of Tregs was partially dependent on prostaglandin E2 (PGE2) produced from SFCs. Moreover, the presence of high CD44+CD24-/low cells in tumor tissues predicted a decreased disease-free survival in patients with EBVaGC. Our study collectively confirmed the existence and immune resistance of CSCs in EBVaGC and offers new insights into the development of novel anti-EBVaGC strategies by targeting CSCs.


Asunto(s)
Antígeno CD24/inmunología , Carcinoma/inmunología , Dinoprostona/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Regulación Neoplásica de la Expresión Génica , Receptores de Hialuranos/inmunología , Neoplasias Gástricas/inmunología , Adulto , Anciano , Animales , Antígeno CD24/genética , Carcinoma/complicaciones , Carcinoma/genética , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Dinoprostona/biosíntesis , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/inmunología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Femenino , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/patogenicidad , Humanos , Receptores de Hialuranos/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Transducción de Señal , Esferoides Celulares/inmunología , Esferoides Celulares/patología , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/virología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Biochem Biophys Res Commun ; 495(2): 1930-1935, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29248729

RESUMEN

Cancer-derived extracellular vesicles (EVs) have emerged as important mediators of tumour-host interactions, and they have been shown to exert various functional effects in immune cells. In most of the studies on human immune cells, EVs have been isolated from cancer cell culture medium or patients' body fluids and added to the immune cell cultures. In such a setting, the physiological relevance of the chosen EV concentration is unknown and the EV isolation method and the timing of EV administration may bias the results. In the current study we aimed to develop an experimental cell culture model to study EV-mediated effects in human T and B cells at conditions mimicking the tumour microenvironment. We constructed a human prostate cancer cell line PC3 producing GFP-tagged EVs (PC3-CD63-GFP cells) and developed a 3D heterotypic spheroid model composed of PC3-CD63-GFP cells and human peripheral blood mononuclear cells (PBMCs). The transfer of GFP-tagged EVs from PC3-CD63-GFP cells to the lymphocytes was analysed by flow cytometry and fluorescence imaging. The endocytic pathway was investigated using three endocytosis inhibitors. Our results showed that GFP-tagged EVs interacted with a large fraction of B cells, however, the majority of EVs were not internalised by B cells but rather remained bound at the cell surface. T cell subsets differed in their ability to interact with the EVs - 15.7-24.1% of the total CD3+ T cell population interacted with GFP-tagged EVs, while only 0.3-5.8% of CD8+ T were GFP positive. Furthermore, a fraction of EVs were internalised in CD3+ T cells via macropinocytosis. Taken together, the heterotypic PC3-CD63-GFP and PBMC spheroid model provides the opportunity to study the interactions and functional effects of cancer-derived EVs in human immune cells at conditions mimicking the tumour microenvironment.


Asunto(s)
Comunicación Celular/inmunología , Técnicas de Cocultivo/métodos , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/patología , Leucocitos Mononucleares/inmunología , Neoplasias Experimentales/inmunología , Esferoides Celulares/inmunología , Línea Celular Tumoral , Humanos , Leucocitos Mononucleares/patología , Neoplasias Experimentales/patología , Esferoides Celulares/patología
7.
J Cell Mol Med ; 21(8): 1503-1512, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28296006

RESUMEN

Cardiosphere-derived cells (CDCs) have been shown to reduce scar size and increase viable myocardium in human patients with mild/moderate myocardial infarction. Studies in rodent models suggest that CDC therapy may confer therapeutic benefits in patients with non-ischaemic dilated cardiomyopathy (DCM). We sought to determine the safety and efficacy of allogeneic CDC in a large animal (canine) model of spontaneous DCM. Canine CDCs (cCDCs) were grown from a donor dog heart. Similar to human CDCs, cCDCs express CD105 and are slightly positive for c-kit and CD90. Thirty million of allogeneic cCDCs was infused into the coronary vessels of Doberman pinscher dogs with spontaneous DCM. Adverse events were closely monitored, and cardiac functions were measured by echocardiography. No adverse events occurred during and after cell infusion. Histology on dog hearts (after natural death) revealed no sign of immune rejection from the transplanted cells.


Asunto(s)
Cardiomiopatía Dilatada/terapia , Vasos Coronarios/citología , Esferoides Celulares/citología , Trasplante de Células Madre , Células Madre/citología , Animales , Biomarcadores/metabolismo , Cardiomiopatía Dilatada/diagnóstico por imagen , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/patología , Vasos Coronarios/inmunología , Perros , Ecocardiografía , Endoglina/genética , Endoglina/inmunología , Femenino , Expresión Génica , Humanos , Masculino , Miocardio/inmunología , Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/inmunología , Esferoides Celulares/inmunología , Esferoides Celulares/trasplante , Células Madre/inmunología , Antígenos Thy-1/genética , Antígenos Thy-1/inmunología , Trasplante Homólogo
8.
Scand J Immunol ; 85(3): 211-219, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27943380

RESUMEN

MCP-1/IL-6 in vitro monocyte secretion upon coculture with autologous fragment spheroids was studied in relation to patient 5- and 10-year overall survival rates in head and neck squamous cell carcinoma (HNSCC) patients (n = 65) diagnosed between 1998 and 2005, nine of whom had an human papilloma virus (HPV) tumour infection. The spheroids were harvested from malignant or benign tissue during primary surgery. Two weeks following surgery, freshly isolated autologous monocytes and benign or malignant spheroids were cocultured 24 h in vitro. The IL-6 secretion was expressed as a fraction of the lipopolysaccharide (LPS) response from the same batch of monocytes. HPV status was obtained by employing PCR analyses of primary diagnostic blocks. IL-6/MCP-1 response levels were not found to be dependent on HPV infection status. MCP-1 secretion did not predict prognosis, nor did in vitro IL-6 monocyte background or LPS-stimulated IL-6 secretion. At 5-year observation, dichotomized IL-6 levels following monocyte coculture, with both malignant and benign spheroids, showed a strong trend towards predicting survival, that is a low monocyte malignant coculture response showed a survival of 31 ± 17 versus 58 ± 17% with a high such response (P = 0.057). When studying monocyte IL-6 coculture responses evaluating benign and malignant spheroid results statistically together, a prediction of survival up to 10 years was found (hazard ratio = 0.48; confidence interval = 0.24-0.96; P < 0.05) with double low IL-6 responses. This survival prediction was also present after an adjustment for HPV tumour infection status. In conclusion, monocyte IL-6 in vitro secretion in cocultures with autologous spheroids/serum from HNSCCs predicted 5- and 10-year survivals, both with and without tumour HPV tumour adjustment.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Quimiocina CCL2/metabolismo , Neoplasias de Cabeza y Cuello/inmunología , Interleucina-6/metabolismo , Monocitos/inmunología , Membrana Mucosa/inmunología , Esferoides Celulares/inmunología , Carcinoma de Células Escamosas/mortalidad , Técnicas de Cocultivo , Neoplasias de Cabeza y Cuello/mortalidad , Humanos , Lipopolisacáridos/inmunología , Membrana Mucosa/citología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/virología , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello , Células Tumorales Cultivadas
9.
BMC Cancer ; 15: 956, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26673159

RESUMEN

BACKGROUND: As ovarian cancer stem cells (CSCs) are responsible for tumor initiation, invasion, metastasis, and chemo-resistance, new stratagems that selectively target ovarian CSCs are critically significant. Our previous work have demonstrated that ovarian cancer spheroid cells are tumorigenic and chemo-resistant, and have the properties of ovarian CSCs. Herein, we hypothesized that expressing α-gal epitopes on ovarian spheroid cells may help eliminate CSCs and improve the outcome of therapeutic intervention for ovarian cancer patients. METHODS: Lentivirus-mediated transfer of a pig α(1,3)galactosyltransferase [α1,3GT] enzyme gene into human ovarian cell line SKOV3 cells formed α-gal epitope-expressing cells (SKOV3-gal cells), and then these cells were maintained in a serum-free culture system to form SKOV3-gal spheroid cells. Efficacy of this cell vaccine was demonstrated in α1,3GT knockout mice (α1,3GT KO mice). RESULTS: The antibody titers to α-gal epitopes measured by ELISA were significantly increased in α1,3GT KO mice after immunization with SKOV3-gal spheroid cells. Furthermore, compared with the non-immunized KO mice, the SKOV3 tumors grafted under renal capsules of KO mice immunized with SKOV3-gal spheroid cells grew slower and began to shrink on day 12. Western blot analysis also showed that immunized KO mice can produce effective antibody against certain tumor associated antigens (TAAs) derived from both SKOV3 cells and SKOV3 spheroid cells. The TAAs were further investigated by mass spectrometry and RNA interference (RNAi) technology. The results suggested that antibodies responding to protein c-erbB-2 may be raised in the sera of the mice after immunization with SKOV3-gal spheroid cells. Ultimately, vaccination with SKOV3-gal spheroid cells induced more CD3+CD4+T cells in the spleen of immunized mice than non-immunized KO mice. CONCLUSIONS: The results suggest that vaccination using ovarian cancer stem-like cells engineered to express α-gal epitopes may be a novel strategy for treatment of ovarian cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Galactosiltransferasas/inmunología , Neoplasias Glandulares y Epiteliales/inmunología , Células Madre Neoplásicas/inmunología , Neoplasias Ováricas/inmunología , Animales , Western Blotting , Carcinoma Epitelial de Ovario , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Femenino , Citometría de Flujo , Humanos , Espectrometría de Masas , Ratones , Ratones Noqueados , Ratones Desnudos , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Esferoides Celulares/inmunología , Porcinos , Transfección , Células Tumorales Cultivadas
10.
Methods Cell Biol ; 189: 97-115, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39393889

RESUMEN

Adoptive natural killer (NK) cell-based immunotherapy is a promising treatment approach in cancer that is showing notable efficacy against hematological malignancies. However, the success of NK cell immunotherapy in patients with solid tumors is limited due to several barriers, which include the immunosuppressive tumor microenvironment (TME), heterogeneity of tumor cells and poor NK cell infiltration into the tumor. Advances in 3D in vitro culture technologies have opened new avenues for the development of more physiological human cancer models that mimic important tumor features which are absent in traditional 2D studies and may be essential for the improvement of immunotherapies against solid tumors. Here, we describe a comprehensive protocol to generate tumor spheroids from the A549 lung carcinoma cell line, then establish co-cultures with NK cells to, ultimately, determine NK cell functional response with a degranulation assay, a surrogate of NK cell cytotoxicity against tumor spheroids. Additionally, we studied degranulation by stimulating NK cell antibody-dependent cell-mediated cytotoxicity (ADCC) with cetuximab, an IgG1 monoclonal antibody used in cancer therapy. Likewise, other monoclonal antibodies or combination treatments could also be studied in this 3D co-culture system, providing very valuable information to define effective combinations of therapeutic agents able to generate NK cells with high cytotoxic potential that could lead to more successful adoptive NK cell-based therapies for the treatment of solid tumors.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Degranulación de la Célula , Técnicas de Cocultivo , Células Asesinas Naturales , Esferoides Celulares , Humanos , Células Asesinas Naturales/inmunología , Esferoides Celulares/inmunología , Técnicas de Cocultivo/métodos , Células A549 , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Microambiente Tumoral/inmunología , Cetuximab/farmacología
11.
Drug Discov Today ; 29(9): 104114, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39067612

RESUMEN

Three-dimensional (3D) cell culture techniques, which are superior to 2D methods in viability and functionality, are being used to develop innovative cancer vaccines. Tumor spheroids, which are structurally and functionally similar to actual tumors, can be developed using 3D cell culture. These spheroid vaccines have shown superior antitumor immune responses to 2D cell-based vaccines. Dendritic cell vaccines can also be produced more efficiently using 3D cell culture. Personalized cancer vaccines are being developed using 3D cell culture, providing substantial benefits over 2D methods. The more natural conditions of 3D cell culture might promote the expression of tumor antigens not expressed in 2D culture, potentially allowing for more targeted vaccines by co-culturing tumor cells with other cell types. Advanced cancer vaccines using 3D cell cultures are expected soon.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Vacunas contra el Cáncer/inmunología , Humanos , Animales , Neoplasias/inmunología , Desarrollo de Vacunas , Técnicas de Cultivo Tridimensional de Células/métodos , Esferoides Celulares/inmunología , Células Dendríticas/inmunología , Técnicas de Cultivo de Célula/métodos
12.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1523-1535, 2024 May 25.
Artículo en Zh | MEDLINE | ID: mdl-38783813

RESUMEN

The adoptive immunotherapy mediated by tumor-infiltrating lymphocytes (TILs) has shown definite efficacy against various solid tumors. However, the inefficiency of the conventional method based on in vitro expansion of TILs fails to achieve the cell count and high tumor-killing activity required for therapeutic purposes. This study investigated the effect of 3D tumor spheroids on the activation and expansion of TILs in vitro, aiming to provide a novel approach for the expansion of TILs. We procured TILs and primary tumor cells from surgical samples of lung cancer patients and then compared the impacts of lung cancer cell line NCI-H1975 and primary lung cancer cells cultured under 2D and 3D conditions on the activation, expansion, and anti-tumor activity of TILs. Furthermore, we added the programmed cell death protein 1 (PD-1) antibody into the co-culture of primary tumor cells and TILs within a 3D environment to assess the effects of the antibody on TILs. The results showed that compared with 2D cultured tumor cells, the 3D cultured H1975 cells significantly enhanced the expansion of TILs, increasing the proportion of CD3+/CD8+ cells in TILs to 61.6%. The 3D primary tumor model also enhanced the proportion of CD3+/CD8+ cells in TILs (45.5%, 54.4%), induced apoptosis of tumor epithelial cells and decreased the overall tumor cells survival rate (16.7%) after co-culture. PD-1 antibodies further improved the in vitro expansion capacity of TILs mediated by 3D tumor spheroids, resulting in the proportions of 50.9% and 57.0% for CD3+/CD8+ cells and enhancing the antitumor activity significantly (reducing the overall tumor survival rate to 9.36%). In summary, the use of 3D tumor spheroids significantly promoted the expansion and improved the anti-tumor effect of TILs, and the use of the PD-1 antibody further promoted the expansion and tumor-killing effect of TILs.


Asunto(s)
Neoplasias Pulmonares , Linfocitos Infiltrantes de Tumor , Esferoides Celulares , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Esferoides Celulares/inmunología , Línea Celular Tumoral , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Receptor de Muerte Celular Programada 1/inmunología , Inmunoterapia Adoptiva , Técnicas de Cocultivo , Técnicas de Cultivo de Célula , Células Tumorales Cultivadas , Proliferación Celular
13.
Commun Biol ; 7(1): 1302, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39390143

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapies have shown remarkable results in patients with hematological malignancies. However, their success in treating solid tumors has been limited. As an alternative candidate for the CAR therapy, CAR-macrophages (CAR-M) have demonstrated activation and phagocytosis directed by tumor-associated antigen (TAA), showing promise in the treatment of solid tumors. Nevertheless, the mechanisms by which CARs direct tumor chemotaxis and invasion of CAR-M remain poorly understood. In this study, we aim to investigate the role of CARs in CAR-M attachment and infiltration using 3D tumor spheroids, which were created by utilizing a novel self-assembling nucleic acid nanostructure decorated living cells (NAC). Our results demonstrated that CAR-M exhibited higher invasion and killing capacity in 2D model and 3D tumor spheroids. In summary, the 3D NAC assembled tumor spheroid model provides a suitable platform for target screening and pharmacodynamic evaluation of CAR-M.


Asunto(s)
Macrófagos , Receptores Quiméricos de Antígenos , Esferoides Celulares , Humanos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Esferoides Celulares/inmunología , Macrófagos/inmunología , ADN/inmunología , ADN/química , Línea Celular Tumoral , Inmunoterapia Adoptiva/métodos , Nanoestructuras/química
14.
Cancer Res ; 84(15): 2432-2449, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38819641

RESUMEN

In vitro preclinical testing of chimeric antigen receptor (CAR) T cells is mostly carried out in monolayer cell cultures. However, alternative strategies are needed to take into account the complexity and the effects of the tumor microenvironment. Here, we describe the modulation of CAR T-cell activity by malignant cells and fibroblasts in human three-dimensional (3D) in vitro cell models of increasing complexity. In models combining mucin-1 (MUC1) and TnMUC1 CAR T cells with human high-grade serous ovarian cancer cell spheroids, malignant cell-intrinsic resistance to CAR T-cell killing was due to defective death receptor signaling involving TNFα. Adding primary human fibroblasts to spheroids unexpectedly increased the ability of CAR T cells to kill resistant malignant cells as CCL2 produced by fibroblasts activated CCR2/4+ CAR T cells. However, culturing malignant cells and fibroblasts in collagen gels engendered production of a dense extracellular matrix that impeded CAR T-cell activity in a TGFß-dependent manner. A vascularized microfluidic device was developed that allowed CAR T cells to flow through the vessels and penetrate the gels in a more physiological way, killing malignant cells in a TNFα-dependent manner. Complex 3D human cell models may provide an efficient way of screening multiple cytotoxic human immune cell constructs while also enabling evaluation of mechanisms of resistance involving cell-cell and cell-matrix interactions, thus accelerating preclinical research on cytotoxic immune cell therapies in solid tumors. Significance: Three-dimensional in vitro models of increasing complexity uncover mechanisms of resistance to CAR T cells in solid tumors, which could help accelerate development of improved CAR T-cell constructs.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias Ováricas , Receptores Quiméricos de Antígenos , Esferoides Celulares , Linfocitos T , Microambiente Tumoral , Humanos , Femenino , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Inmunoterapia Adoptiva/métodos , Microambiente Tumoral/inmunología , Esferoides Celulares/inmunología , Linfocitos T/inmunología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Línea Celular Tumoral
15.
J Vis Exp ; (206)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38738886

RESUMEN

Monoclonal antibody-based immunotherapy targeting tumor antigens is now a mainstay of cancer treatment. One of the clinically relevant mechanisms of action of the antibodies is antibody-dependent cellular cytotoxicity (ADCC), where the antibody binds to the cancer cells and engages the cellular component of the immune system, e.g., natural killer (NK) cells, to kill the tumor cells. The effectiveness of these therapies could be improved by identifying adjuvant compounds that increase the sensitivity of the cancer cells or the potency of the immune cells. In addition, undiscovered drug interactions in cancer patients co-medicated for previous conditions or cancer-associated symptoms may determine the success of the antibody therapy; therefore, such unwanted drug interactions need to be eliminated. With these goals in mind, we created a cancer ADCC model and describe here a simple protocol to find ADCC-modulating drugs. Since 3D models such as cancer cell spheroids are superior to 2D cultures in predicting in vivo responses of tumors to anticancer therapies, spheroid co-cultures of EGFP-expressing HER2+ JIMT-1 breast cancer cells and the NK92.CD16 cell lines were set up and induced with Trastuzumab, a monoclonal antibody clinically approved against HER2-positive breast cancer. JIMT-1 spheroids were allowed to form in cell-repellent U-bottom 96-well plates. On day 3, NK cells and Trastuzumab were added. The spheroids were then stained with Annexin V-Alexa 647 to measure apoptotic cell death, which was quantitated in the peripheral zone of the spheroids with an automated microscope. The applicability of our assay to identify ADCC-modulating molecules is demonstrated by showing that Sunitinib, a receptor tyrosine kinase inhibitor approved by the FDA against metastatic cancer, almost completely abolishes ADCC. The generation of the spheroids and image acquisition and analysis pipelines are compatible with high-throughput screening for ADCC-modulating compounds in cancer cell spheroids.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Esferoides Celulares , Humanos , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/inmunología , Descubrimiento de Drogas/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de los fármacos , Línea Celular Tumoral , Receptores de IgG/inmunología , Antineoplásicos Inmunológicos/farmacología , Trastuzumab/farmacología
16.
J Immunol ; 187(7): 3806-14, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21880978

RESUMEN

Tumor-associated macrophages are known to amplify the malignant potential of tumors by secreting a variety of cytokines and proteases involved in tumor cell invasion and metastasis, but how these macrophages infiltrate tumors and whether the macrophage migration process facilitates tumor cell invasion remain poorly documented. To address these questions, we used cell spheroids of breast carcinoma SUM159PT cells as an in vitro model of solid tumors. We found that macrophages used both the mesenchymal mode requiring matrix metalloproteinases (MMPs) and the amoeboid migration mode to infiltrate tumor cell spheroids. Whereas individual SUM159PT cells invaded Matrigel using an MMP-dependent mesenchymal mode, when they were grown as spheroids, tumor cells were unable to invade the Matrigel surrounding spheroids. When spheroids were infiltrated or in contact with macrophages, tumor cell invasiveness was restored. It was dependent on the capacity of macrophages to remodel the matrix and migrate in an MMP-independent mesenchymal mode. This effect of macrophages was much reduced when spheroids were infiltrated by Matrigel migration-defective Hck(-/-) macrophages. In the presence of macrophages, SUM159PT migrated into Matrigel in the proximity of macrophages and switched from an MMP-dependent mesenchymal migration to an amoeboid mode resistant to protease inhibitors.Thus, in addition to the well-described paracrine loop between macrophages and tumor cells, macrophages can also contribute to the invasiveness of tumor cells by remodeling the extracellular matrix and by opening the way to exit the tumor and colonize the surrounding tissues in an MMP-dispensable manner.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Macrófagos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Invasividad Neoplásica/inmunología , Esferoides Celulares/inmunología , Animales , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Macrófagos/inmunología , Macrófagos/ultraestructura , Ratones , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Invasividad Neoplásica/patología , Esferoides Celulares/metabolismo , Esferoides Celulares/ultraestructura
17.
J Immunol ; 186(3): 1538-45, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21191066

RESUMEN

NK cell activation is negatively regulated by the expression of target cell MHC class I molecules. We show that this relationship is nonlinear due to an NK cell activation/inhibition threshold. Ewing's sarcoma family tumor cell monolayers, which were highly susceptible to NK cells in vitro, developed a highly resistant phenotype when cultured as three-dimensional multicellular tumor spheroid structures. This suggested that tumor architecture is likely to influence the susceptibility to NK cells in vivo. Resistance of the multicellular tumor spheroid was associated with the increased expression of MHC class I molecules and greatly reduced NK cell activation, implying that a threshold of NK cell activation/inhibition had been crossed. Reducing MHC class I expression on Ewing's sarcoma family tumor monolayers did not alter their susceptibility to NK cells, whereas increased expression of MHC class I rendered them resistant and allowed the threshold point to be identified. This threshold, as defined by MHC class I expression, was predictive of the number of NK-resistant target cells within a population. A threshold permits modest changes in the target cell surface phenotype to profoundly alter the susceptibility to NK cells. Whereas this allows for the efficient detection of target cells, it also provides a route for pathogens and tumors to evade NK cell attack.


Asunto(s)
Membrana Celular/inmunología , Predisposición Genética a la Enfermedad/genética , Células Asesinas Naturales/inmunología , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/metabolismo , Escape del Tumor/inmunología , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/patología , Pruebas Inmunológicas de Citotoxicidad/métodos , Células HeLa , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inmunofenotipificación/métodos , Células K562 , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Activación de Linfocitos/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Sarcoma de Ewing/patología , Esferoides Celulares/inmunología , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Translocación Genética/inmunología , Células Tumorales Cultivadas , Escape del Tumor/genética
18.
Tsitologiia ; 55(1): 36-44, 2013.
Artículo en Ruso | MEDLINE | ID: mdl-23662577

RESUMEN

Two continuous cell lines, monolayer (ZH-ad) and suspension (ZH-fl), which doffer in the level of cell differentiation and tumorigenicity, were obtained from explants of ascitic Zajdela hepatoma. Using tumor-specific rabbit immune serum tumor-associated antigens were detected in the fraction of plasma membrane of ZH-fl cells, synthesis of that was reduced or suppressed in ZH-ad cells. Intraperitoneal injection of ZH-fl cells as few as 0.5 - 10(6) cells/rat was enough for development of ascites tumor, while minimal dose for ZH-ad cells increase to 20 - 10(6) cells. Three types of generated clones were revealed by means of clonogenic analysis of ZH-fl cells: nonadhesive sphere colonies and two types of monolayer clones ranging in proliferative potential, shape of colonies and cell composition. When the spherical colonies had reached a critical size they disintegrated with dissociation of single cells or islets of various sizes, part of which attached to the plate to form a monolayer. Three clonal cell lines were obtained: IC - as a result of expansion of sphere clone cells, 4G and 10E - from monolayer clones. Our results demonstrate a clonogenic origin of multicellular islets of ascetic hepatoma Zajdela.


Asunto(s)
Antígenos de Neoplasias , Carcinoma Hepatocelular , Neoplasias Hepáticas , Esferoides Celulares , Animales , Anticuerpos Antineoplásicos/inmunología , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/inmunología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Conejos , Ratas , Esferoides Celulares/inmunología , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
19.
Br J Cancer ; 106(6): 1123-33, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22374462

RESUMEN

BACKGROUND: Novel treatment strategies are needed to cure disseminated Ewing sarcoma. Primitive neuroectodermal features and a mesenchymal stem cell origin are both compatible with aberrant expression of the ganglioside antigen G(D2) and led us to explore G(D2) immune targeting in this cancer. METHODS: We investigated G(D2) expression in Ewing sarcoma by immunofluorescence staining. We then assessed the antitumour activity of T cells expressing a chimeric antigen receptor specific for G(D2) against Ewing sarcoma in vitro and in vivo. RESULTS: Surface G(D2) was detected in 10 out of 10 Ewing sarcoma cell lines and 3 out of 3 primary cell cultures. Moreover, diagnostic biopsies from 12 of 14 patients had uniform G(D2) expression. T cells specifically modified to express the G(D2)-specific chimeric receptor 14. G2a-28ζ efficiently interacted with Ewing sarcoma cells, resulting in antigen-specific secretion of cytokines. Moreover, chimeric receptor gene-modified T cells from healthy donors and from a patient exerted potent, G(D2)-specific cytolytic responses to allogeneic and autologous Ewing sarcoma, including tumour cells grown as multicellular, anchorage-independent spheres. G(D2)-specific T cells further had activity against Ewing sarcoma xenografts. CONCLUSION: G(D2) surface expression is a characteristic of Ewing sarcomas and provides a suitable target antigen for immunotherapeutic strategies to eradicate micrometastatic cells and prevent relapse in high-risk disease.


Asunto(s)
Neoplasias Óseas/metabolismo , Gangliósidos/metabolismo , Sarcoma de Ewing/metabolismo , Linfocitos T/trasplante , Adolescente , Adulto , Animales , Antígenos de Superficie/inmunología , Antígenos de Superficie/metabolismo , Neoplasias Óseas/inmunología , Neoplasias Óseas/terapia , Línea Celular Tumoral , Proliferación Celular , Niño , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Femenino , Gangliósidos/inmunología , Granzimas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Receptores de Antígenos de Linfocitos T/biosíntesis , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/terapia , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/metabolismo , Esferoides Celulares/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto Joven
20.
Breast Cancer Res Treat ; 134(1): 61-70, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22173696

RESUMEN

Cancer progenitor cells are critical for tumor initiation and recurrence so they are an important therapeutic target. We tested whether T cells could recognize tumor antigens expressed by breast cancer progenitor cells and acquire therapeutic activity against established metastases or delay onset of spontaneous tumors. Breast tumors were derived from HER2/neu transgenic mice and propagated in vitro under conditions that selected progenitor cells which were then used as an irradiated whole cell vaccine. A minor subset of recently sensitized T cells was isolated from vaccine-draining lymph nodes then activated in vitro to achieve numerical expansion. We show that the tumor progenitor cell vaccines reversed tolerance to a known HER2/neu epitope, otherwise inhibited by Treg cells. Additional shared tumor antigens were recognized because a Neuneg subclone also induced a Th1 type immune response against breast tumors. Adoptive transfer of in vitro activated lymph node T cells-mediated regression of established metastases from multiple independently derived breast tumor lines. Moreover, adoptive transfer of effector T cells into Neu-tolerant mice, months before the onset of spontaneous tumors, significantly postponed tumor development. Interestingly, T-cell-mediated lysis of metastases stimulated an IgG response to HER2/neu as well as other shared antigens. In summary, tumor progenitor cells contain shared antigens which can lead to a cross-protective T-cell response. Moreover, antigens acquired during immune-mediated tumor destruction are presented in a manner conducive to reversal of tolerance and Ig class switching. These complementary effector mechanisms might augment therapy by eliminating refractory breast cancer stem cells.


Asunto(s)
Carcinoma/terapia , Inmunoterapia Adoptiva , Neoplasias Mamarias Experimentales/terapia , Células Madre Neoplásicas/inmunología , Receptor ErbB-2/genética , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer , Carcinoma/genética , Carcinoma/secundario , Femenino , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Receptor ErbB-2/inmunología , Esferoides Celulares/inmunología , Linfocitos T/inmunología , Linfocitos T/trasplante , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA