Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 11(1): 2014655, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36524207

RESUMEN

Clonal cell line-based, multigene-modified, off-the-shelf NK cell therapeutics are emerging as the new frontier of adoptive cellular immunotherapy. Here, we utilized a newly established NK cell line, NK101, as a backbone to derive multifaceted killer cells armored with various antitumor modalities through repeated cycles of genetic modification and clonal selection. First, NK101 cells were transduced with a tricistronic lentiviral vector expressing CD7, CD28, and cytosine deaminase (CD). The resulting cell line demonstrated enhanced cytotoxicity against B7+ tumors and exerted bystander killing effects on neighboring tumor cells upon 5-FC treatment. Second, engineered NK101 cells were again transduced with a bicistronic vector expressing membrane-bound interleukin-15 (mbIL-15) and dominant negative TGFß type II receptor (DNTßRII). Ectopic expression of mbIL-15 resulted in further augmentation of lytic activities against all tested target cells by inducing upregulation of multiple activating receptors, while that of DNTßRII allowed the cells to maintain heightened cytotoxicity in the presence of TGFß. Finally, dual-transduced NK101 cells were modified to express chimeric antigen receptors (CARs) targeting either a solid tumor antigen (EpCAM) or a hematologic tumor antigen (FLT3). The final engineered products not only demonstrated antigen-specific killing activities in vitro but also exerted strong tumor-inhibitory effects in preclinical models of metastatic solid tumor and hematologic malignancy. Notably, combined treatment with 5-FC further enhanced antitumor efficacy of engineered NK101 in the solid tumor model. Our results demonstrate successful generation of multigene-modified NK101 cell therapeutics exerting diverse mechanisms of antitumor action - activation receptor-mediated innate killing, antigen-specific killing, and bystander effect-mediated killing.


Asunto(s)
Citotoxicidad Inmunológica , Células Asesinas Naturales , Línea Celular Tumoral , Células Asesinas Naturales/metabolismo , Inmunoterapia Adoptiva/métodos , Factor de Crecimiento Transformador beta/metabolismo
2.
J Immunother Cancer ; 7(1): 138, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31126350

RESUMEN

BACKGROUND: Human natural killer (NK) cell lines serve as an attractive source for adoptive immunotherapy, but NK-92 remains the only cell line being assessed in the clinic. Here, we established a novel NK cell line, NK101, from a patient with extra-nodal natural killer/T-cell lymphoma and examined its phenotypic, genomic and functional characteristics. METHODS: Single cell suspensions from lymphoma tissue were expanded with anti-NKp46/anti-CD2-coated beads in the presence of IL-2. A continuously growing CD56+ cell clone was selected and designated as NK101. Flow cytometry and RNA sequencing were used to characterize phenotypic and genomic features of NK101. In vitro cytotoxicity and IFN-γ/TNF-α secretion were measured by flow cytometry-based cytotoxicity assay and enzyme-linked immunosorbent assay, respectively, after direct co-culture with tumor cells. Immunomodulatory potential of NK101 was assessed in an indirect co-culture system using conditioned medium. Finally, in vivo antitumor efficacy was evaluated in an immunocompetent, syngeneic 4T1 mammary tumor model. RESULTS: NK101 displayed features of CD56dimCD62L+ intermediate stage NK subset with the potential to simultaneously act as a cytokine producer and a cytotoxic effector. Comparative analysis of NK101 and NK-92 revealed that NK101 expressed lower levels of perforin and granzyme B that correlated with weaker cytotoxicity, but produced higher levels of pro-inflammatory cytokines including IFN-γ and TNF-α. Contrarily, NK-92 produced greater amounts of anti-inflammatory cytokines, IL-1 receptor antagonist and IL-10. Genome-wide analysis revealed that genes associated with positive regulation of leukocyte proliferation were overexpressed in NK101, while those with opposite function were highly enriched in NK-92. The consequence of such expressional and functional discrepancies was well-represented in (i) indirect co-culture system where conditioned medium derived from NK101 induced greater proliferation of human peripheral blood mononuclear cells and (ii) immunocompetent 4T1 tumor model where peritumoral injections of NK101 displayed stronger anti-tumor activities by inducing higher tumor-specific immune responses. In a manufacturing context, NK101 not only required shorter recovery time after thawing, but also exhibited faster growth profile than NK-92, yielding more than 200-fold higher cell numbers after 20-day culture. CONCLUSION: NK101 is a unique NK cell line bearing strong immunostimulatory potential and substantial scalability, providing an attractive source for adoptive cancer immunotherapy.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Inmunoterapia/métodos , Traslado Adoptivo , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Neoplasias
3.
Immune Netw ; 17(5): 343-351, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29093655

RESUMEN

Developing a novel vaccine that can be applied against multiple strains of influenza virus is of utmost importance to human health. Previously, we demonstrated that the intranasal introduction of Fc-fused IL-7 (IL-7-mFc), a long-acting cytokine fusion protein, confers long-lasting prophylaxis against multiple strains of influenza A virus (IAV) by inducing the development of lung-resident memory-like T cells, called TRM-like cells. Here, we further investigated the mechanisms of IL-7-mFc-mediated protective immunity to IAVs. First, we found that IL-7-mFc treatment augments the accumulation of pulmonary T cells in 2 ways: recruiting blood circulating T cells into the lung and expanding T cells at the lung parenchyma. Second, the blockade of T cell migration from the lymph nodes (LNs) with FTY720 treatment was not required for mounting the protective immunity to IAV with IL-7-mFc, suggesting a more important role of IL-7 in T cells in the lungs. Third, IL-7-mFc treatment also recruited various innate immune cells into the lungs. Among these cells, plasmacytoid dendritic cells (pDCs) play an important role in IL-7-mFc-mediated protective immunity through reducing the immunopathology and increasing IAV-specific cytotoxic T lymphocyte (CTL) responses. In summary, our results show that intranasal treatment with IL-7-mFc modulates pulmonary immune responses to IAV, affecting both innate and adaptive immune cells.

4.
Nanoscale ; 10(1): 150-157, 2017 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-29115327

RESUMEN

On-the-spot visualization of biochemical responses of intact live cells is vital for a clear understanding of cell biology. The main obstacles for instant visualization of biochemical responses of living cells arise from the lack of a sophisticated detecting technique which can simultaneously provide chemical analysis tools and the biocompatible wet conditions. Here we introduce scanning transmission X-ray microscopy (STXM) combined with a liquid-enclosing graphene system (LGS), offering biocompatible conditions and improved X-ray absorption spectra to probe the chemical responses of live cells under wet conditions. This set-up enables us to probe a subtle change in absorption spectra depending on the oxidation state of a miniscule amount of oxygen in the functional groups present in each cell and its surroundings containing a minimal amount of liquid water. As an example of in situ biochemical responses of wet cells, chemical responses of a single Colo 205 cell are visualized and analyzed using X-ray absorption near the oxygen K-edge. This spectromicroscopic method using LGS can be applied to diverse biological samples under wet conditions for the analysis of their biochemical responses.


Asunto(s)
Grafito/química , Microscopía/métodos , Análisis Espectral/métodos , Materiales Biocompatibles , Línea Celular , Humanos , Microscopía Confocal , Oxidación-Reducción , Oxígeno , Agua , Rayos X
6.
Clin Cancer Res ; 22(23): 5898-5908, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27407095

RESUMEN

PURPOSE: The induction of tissue-localized virus-specific CD8 T-cell response is essential for the development of an effective therapeutic vaccine against genital diseases, such as cervical cancer and genital herpes. Here, we aimed to elucidate the immunologic role of IL7 in the induction of mucosal cellular immunity. EXPERIMENTAL DESIGN: IL7 was engineered through Fc fusion to enhance mucosal delivery across the genital epithelial barrier. The immunomodulatory role of IL7 was evaluated by monitoring the kinetics of various immune cells and measuring the expression of chemokines and cytokines after intravaginal administration of Fc-fused IL7 (IL7-Fc). The antitumor effects of intramuscular human papillomavirus (HPV) DNA vaccine or topical IL7-Fc alone or in a combinational regimen on mice survival were compared using a orthotopic cervical cancer model. RESULTS: Intravaginal treatment of IL7-Fc, but not native IL7, induces upregulation of chemokines (CXCL10, CCL3, CCL4, and CCL5), cytokines (IFNγ, TNFα, IL6, and IL1ß), and an adhesion molecule (VCAM-1) in the genital tract, leading to the recruitment of several leukocytes, including CD4, CD8, γδ T cells, and dendritic cells. Importantly, in this murine cervical cancer model, topical administration of IL7-Fc after intramuscular HPV DNA vaccination increases the number of HPV-specific CD8 T cells in the genital mucosa, but not in the spleen, leading to stronger antitumor activity than the HPV DNA vaccine alone. CONCLUSIONS: Our findings provide an important insight into the immunomodulatory role of IL7-Fc via topical application and the design of therapeutic vaccine regimen that induces effective genital-mucosal CD8 T-cell responses. Clin Cancer Res; 22(23); 5898-908. ©2016 AACR.


Asunto(s)
Alphapapillomavirus/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Interleucina-7/administración & dosificación , Interleucina-7/inmunología , Neoplasias del Cuello Uterino/inmunología , Vacunas de ADN/inmunología , Administración Intravaginal , Animales , Citocinas/inmunología , Células Dendríticas/inmunología , Femenino , Inmunidad Celular/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/inmunología , Neoplasias del Cuello Uterino/virología , Vacunación/métodos
7.
J Virol ; 90(5): 2273-84, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26656713

RESUMEN

UNLABELLED: Influenza A virus (IAV) infection frequently causes hospitalization and mortality due to severe immunopathology. Annual vaccination and antiviral drugs are the current countermeasures against IAV infection, but they have a limited efficacy against new IAV variants. Here, we show that intranasal pretreatment with Fc-fused interleukin-7 (IL-7-mFc) protects mice from lethal IAV infections. The protective activity of IL-7-mFc relies on transcytosis via neonatal Fc receptor (FcRn) in the lung and lasts for several weeks. Introduction of IL-7-mFc alters pulmonary immune environments, leading to recruitment of T cells from circulation and their subsequent residency as tissue-resident memory-like T (TRM-like) cells. IL-7-mFc-primed pulmonary TRM-like cells contribute to protection upon IAV infection by dual modes. First, TRM-like cells, although not antigen specific but polyclonal, attenuate viral replication at the early phase of IAV infection. Second, TRM-like cells augment expansion of IAV-specific cytotoxic T lymphocytes (CTLs), in particular at the late phase of infection, which directly control viruses. Thus, accelerated viral clearance facilitated by pulmonary T cells, which are either antigen specific or not, alleviates immunopathology in the lung and mortality from IAV infection. Depleting a subset of pulmonary T cells indicates that both CD4 and CD8 T cells contribute to protection from IAV, although IL-7-primed CD4 T cells have a more prominent role. Collectively, we propose intranasal IL-7-mFc pretreatment as an effective means for generating protective immunity against IAV infections, which could be applied to a potential prophylaxis for influenza pandemics in the future. IMPORTANCE: The major consequence of a highly pathogenic IAV infection is severe pulmonary inflammation, which can result in organ failure and death at worst. Although vaccines for seasonal IAVs are effective, frequent variation of surface viral proteins hampers development of protective immunity. In this study, we demonstrated that intranasal IL-7-mFc pretreatment protected immunologically naive mice from lethal IAV infections. Intranasal pretreatment with IL-7-mFc induced an infiltration of T cells in the lung, which reside as effector/memory T cells with lung-retentive markers. Those IL-7-primed pulmonary T cells contributed to development of protective immunity upon IAV infection, reducing pulmonary immunopathology while increasing IAV-specific cytotoxic T lymphocytes. Since a single treatment with IL-7-mFc was effective in the protection against multiple strains of IAV for an extended period of time, our findings suggest a possibility that IL-7-mFc treatment, as a potential prophylaxis, can be developed for controlling highly pathogenic IAV infections.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Factores Inmunológicos/administración & dosificación , Virus de la Influenza A/inmunología , Interleucina-7/administración & dosificación , Pulmón/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Administración Intranasal , Animales , Femenino , Fragmentos Fc de Inmunoglobulinas/genética , Factores Inmunológicos/genética , Interleucina-7/genética , Depleción Linfocítica , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Linfocitos T/inmunología
8.
J Virol ; 88(16): 8998-9009, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899182

RESUMEN

UNLABELLED: T follicular helper (Tfh) cells are specialized providers of cognate B cell help, which is important in promoting the induction of high-affinity antibody production in germinal centers (GCs). Interleukin-6 (IL-6) and IL-21 have been known to play important roles in Tfh cell differentiation. Here, we demonstrate that IL-7 plays a pivotal role in Tfh generation and GC formation in vivo, as treatment with anti-IL-7 neutralizing antibody markedly impaired the development of Tfh cells and IgG responses. Moreover, codelivery of mouse Fc-fused IL-7 (IL-7-mFc) with a vaccine enhanced the generation of GC B cells as well as Tfh cells but not other lineages of T helper cells, including Th1, Th2, and Th17 cells. Interestingly, a 6-fold-lower dose of an influenza virus vaccine codelivered with Fc-fused IL-7 induced higher antigen-specific and cross-reactive IgG titers than the vaccine alone in both mice and monkeys and led to markedly enhanced protection against heterologous influenza virus challenge in mice. Enhanced generation of Tfh cells by IL-7-mFc treatment was not significantly affected by the neutralization of IL-6 and IL-21, indicating an independent role of IL-7 on Tfh differentiation. Thus, IL-7 holds promise as a critical cytokine for selectively inducing Tfh cell generation and enhancing protective IgG responses. IMPORTANCE: Here, we demonstrate for the first time that codelivery of Fc-fused IL-7 significantly increased influenza virus vaccine-induced antibody responses, accompanied by robust expansion of Tfh cells and GC B cells as well as enhanced GC formation. Furthermore, IL-7-mFc induced earlier and cross-reactive IgG responses, leading to striking protection against heterologous influenza virus challenge. These results suggest that Fc-fused IL-7 could be used for inducing strong and cross-protective humoral immunity against highly mutable viruses, such as HIV and hepatitis C virus, as well as influenza viruses.


Asunto(s)
Inmunidad Humoral/inmunología , Interleucina-7/inmunología , Activación de Linfocitos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Femenino , Centro Germinal/inmunología , Haplorrinos/inmunología , Inmunoglobulina G/inmunología , Vacunas contra la Influenza/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Orthomyxoviridae/inmunología
9.
Oncotarget ; 5(5): 1226-40, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24713374

RESUMEN

Identification of novel biomarkers for tumor-initiating cells (TICs) is of critical importance for developing diagnostic and therapeutic strategies against cancers. Here we identified the role of KIAA1114, a full-length translational product of the trophinin gene, as a distinctive marker for TICs in human liver cancer by developing a DNA vaccine-induced monoclonal antibody targeting the putative extracellular domain of KIAA1114. Compared with other established markers of liver TICs, KIAA1114 was unique in that its expression was detected in both alpha fetoprotein (AFP)-positive and AFP-negative hepatocellular carcinoma (HCC) cell lines with the expression levels of KIAA1114 being positively correlated to their tumorigenic potentials. Notably, KIAA1114 expression was strongly detected in primary hepatic tumor, but neither in the adjacent non-tumorous tissue from the same patient nor normal liver tissue. KIAA1114high cells isolated from HCC cell lines displayed TIC-like features with superior functional and phenotypic traits compared to their KIAA1114low counterparts, including tumorigenic abilities in xenotransplantation model, in vitro colony- and spheroid-forming capabilities, expression of stemness-associated genes, and migratory capacity. Our findings not only address the value of a novel antigen, KIAA1114, as a potential diagnostic factor of human liver cancer, but also as an independent biomarker for identifying TIC populations that could be broadly applied to the heterogeneous HCC subtypes.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Moléculas de Adhesión Celular/metabolismo , Colangiocarcinoma/metabolismo , Neoplasias Hepáticas/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Anticuerpos Monoclonales , Antígenos de Superficie , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Carcinoma Hepatocelular/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Línea Celular Tumoral , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/genética , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/metabolismo , Esferoides Celulares , Ensayo de Tumor de Célula Madre , alfa-Fetoproteínas/metabolismo
10.
Clin Cancer Res ; 19(2): 415-27, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23204131

RESUMEN

PURPOSE: Durable complete remission of metastatic renal cell carcinoma (RCC) has rarely been achieved with current treatment modalities. To solve this problem, alternative therapeutic options with high efficacy and minimal side effects are strongly needed. EXPERIMENTAL DESIGN: Mesenchymal stem cells (MSC) were engineered to coexpress dodecameric TRAIL and herpes simplex virus thymidine kinase (MSC/dTRAIL-TK). The antitumor effects of MSCs expressing dTRAIL (MSC/dTRAIL) or HSV-TK alone (MSC/TK) and MSC/dTRAIL-TK were compared with murine RCC cells using in vitro coculture system and in vivo experimental lung metastasis model. The effects of different doses and schedules of engineered MSCs on mice survival were also evaluated. RESULTS: MSC/dTRAIL-TK exerted stronger apoptotic response in Renca cells than did MSC/TK or MSC/dTRAIL after ganciclovir (GCV) treatment. In vivo imaging results suggest that MSCs reside longer in the lungs of metastatic tumor-bearing mice, compared with that of control mice, regardless of genetic engineering. In addition, MSC/dTRAIL-TK treatment followed by ganciclovir administrations significantly decreased the number of tumor nodules in the lung, to a greater degree than MSC/dTRAIL or MSC/TK, and led to a prolonged survival. More importantly, the antimetastatic effect of MSC/dTRAIL-TK was markedly enhanced by repeated injections but not by increased dose, and resulted in 100% survival of tumor-bearing mice after three injections. CONCLUSION: Sequential combination gene therapy using MSC/dTRAIL-TK achieved long-term remission of metastatic RCC without noticeable toxicity. Our findings provide an innovative therapeutic approach to completely eradicate metastatic tumors by simple, repeated administrations of MSC/dTRAIL-TK.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/secundario , Neoplasias Renales/genética , Neoplasias Renales/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Células Madre Mesenquimatosas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Timidina Quinasa/genética , Animales , Apoptosis/genética , Carcinoma de Células Renales/terapia , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Neoplasias Renales/terapia , Neoplasias Pulmonares/terapia , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratas , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA