Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Health Commun ; : 1-11, 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37415274

RESUMEN

The lack of daily conversation may lead to the deterioration of quality of life and cognitive function in older adults requiring long-term care. This study aimed to develop a scale to measure daily conversation among them, the Life-Worldly Communication Scale: LWCS, and to test its structural, convergent, and discriminant validity. The subjects were 539 older adults requiring long-term care in facilities and at home. A 24-item provisional scale was created, using a panel of experts. Structural validity of LWCS was examined from exploratory factor analysis to establish the factor structure, two confirmatory factor analyses for cross-validation, and measurement invariance between the institutional and home setting. Convergent validity was examined from the average variance extracted: AVE, composite reliability: CR, and simple regression analysis between LWCS and Interdependent Happiness Scale: IHS. Discriminant validity was examined using the heterotrait-monotrait ratio of correlations: HTMT. Multiple imputations were used to deal with missing data on these scales. The results showed that the goodness of fit of the three-factor, 11-item model obtained from the two-step CFA was SRMR = .043, RMSEA = .059, CFI = .978, and AGFI = .905. The model was confirmed for structural validity by measurement invariance tests: configural invariance (CFI=.973, RMSEA = .047), metric invariance (ΔCFI= .001, ΔRMSEA=-.004), scalar invariance (ΔCFI =-0.002, RMSEA = -0.003). Convergent validity was confirmed by AVE = .503~.772, CR = .801~.910, and simple regression analysis between LWCS and IHS (adjusted r2=.18, p < .001). Discriminant validity was also confirmed among the three factors (HTMT=.496~.644). LWCS can contribute to the assessment of daily conversation in geriatric settings and research regarding its promotion.

2.
Angew Chem Int Ed Engl ; 62(8): e202216142, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36562327

RESUMEN

Cytotoxic immune cells, including T lymphocytes (CTLs) and natural killer (NK) cells, are essential components of the host response against tumors. CTLs and NK cells secrete granzyme A (GzmA) upon recognition of cancer cells; however, there are very few tools that can detect physiological levels of active GzmA with high spatiotemporal resolution. Herein, we report the rational design of the near-infrared fluorogenic substrates for human GzmA and mouse GzmA. These activity-based probes display very high catalytic efficiency and selectivity over other granzymes, as shown in tissue lysates from wild-type and GzmA knock-out mice. Furthermore, we demonstrate that the probes can image how adaptive immune cells respond to antigen-driven recognition of cancer cells in real time.


Asunto(s)
Colorantes Fluorescentes , Linfocitos T Citotóxicos , Animales , Humanos , Ratones , Granzimas , Células Asesinas Naturales , Ratones Noqueados
3.
Angew Chem Int Ed Engl ; 61(41): e202207508, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-35993914

RESUMEN

Increased levels of tumor-associated macrophages (TAMs) are indicators of poor prognosis in most cancers. Although antibodies and small molecules blocking the recruitment of macrophages to tumors are under evaluation as anticancer therapies, these strategies are not specific for macrophage subpopulations. Herein we report the first enzyme-activatable chemokine conjugates for effective targeting of defined macrophage subsets in live tumors. Our constructs exploit the high expression of chemokine receptors (e.g., CCR2) and the activity of cysteine cathepsins in TAMs to target these cells selectively over other macrophages and immune cells (e.g., neutrophils, T cells, B cells). Furthermore, we demonstrate that cathepsin-activatable chemokines are compatible with both fluorescent and therapeutic cargos, opening new avenues in the design of targeted theranostic probes for immune cells in the tumor microenvironment.


Asunto(s)
Cisteína , Macrófagos Asociados a Tumores , Catepsinas , Quimiocinas , Receptores de Quimiocina , Microambiente Tumoral
4.
Angew Chem Int Ed Engl ; 60(11): 5699-5703, 2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33300671

RESUMEN

Natural killer (NK) cells are immune cells that can kill certain types of cancer cells. Adoptive transfer of NK cells represents a promising immunotherapy for malignant tumours; however, there is a lack of methods to validate anti-tumour activity of NK cells in vivo. Herein, we report a new chemiluminescent probe to image in situ the granzyme B-mediated killing activity of NK cells against cancer cells. We have optimised a granzyme B-specific construct using an activatable phenoxydioxetane reporter so that enzymatic cleavage of the probe results in bright chemiluminescence. The probe shows high selectivity for active granzyme B over other proteases and higher signal-to-noise ratios than commercial fluorophores. Finally, we demonstrate that the probe can detect NK cell activity in mouse models, being the first chemiluminescent probe for in vivo imaging of NK cell activity in live tumours.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Granzimas/metabolismo , Células Asesinas Naturales/metabolismo , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Colorantes Fluorescentes/química , Granzimas/química , Humanos , Células Asesinas Naturales/patología , Mediciones Luminiscentes , Ratones , Estructura Molecular , Neoplasias/diagnóstico por imagen , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/metabolismo , Imagen Óptica
6.
Angew Chem Int Ed Engl ; 58(47): 16894-16898, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31535788

RESUMEN

We report the novel chemical design of fluorescent activatable chemokines as highly specific functional probes for imaging subpopulations of immune cells in live tumours. Activatable chemokines behave as AND-gates since they emit only after receptor binding and intracellular activation, showing enhanced selectivity over existing agents. We have applied this strategy to produce mCCL2-MAF as the first probe for in vivo detection of metastasis-associated macrophages in a preclinical model of lung metastasis. This strategy will accelerate the preparation of new chemokine-based probes for imaging immune cell function in tumours.


Asunto(s)
Neoplasias de la Mama/patología , Colorantes Fluorescentes/química , Neoplasias Pulmonares/patología , Macrófagos/patología , Imagen Molecular/métodos , Receptores CCR2/fisiología , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espectrometría de Fluorescencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Immunology ; 155(3): 285-293, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29963704

RESUMEN

Tumour-infiltrating immune cells regulate tumour development and progression either negatively or positively. For example, cytotoxic lymphocytes (CTL) such as CD8+ T and natural killer (NK) cells can recognize and eliminate cancer cells, and thereby restrict the tumour growth and metastasis, if they exert full cytotoxicity. In contrast, tumour-infiltrating myeloid cells such as tumour-associated macrophages (TAM) promote the expansion and dissemination of cancer cells depending on their functional states. Given the tumour-killing ability of CTL, the augmentation of CTL-induced antitumour immune reactions has been considered as an attractive therapeutic modality for lethal solid tumours and several promising strategies have emerged, which include immune checkpoint inhibitors, cancer vaccines and adoptive CTL transfer. These immunotherapies are now tested in clinical trials and have shown significant antitumour effects in patients with lymphoma and some solid tumours such as melanoma and lung cancer. Despite these encouraging results, these therapies are not efficient in a certain fraction of patients and tumour types with tumour cell-intrinsic mechanisms such as impaired antigen presentation and/or tumour cell-extrinsic mechanisms including the accumulation of immunosuppressive cells. Several animal studies suggest that tumour-infiltrating myeloid cells, especially TAM, are one of the key targets to improve the efficacy of immunotherapies as these cells can suppress the functions of CD8+ T and NK cells. In this review, we will summarize recent animal studies regarding the involvement of TAM in the immune checkpoint, cancer vaccination and adoptive CTL transfer therapies, and discuss the therapeutic potential of TAM targeting to improve the immunotherapies.


Asunto(s)
Inmunoterapia , Neoplasias Pulmonares , Macrófagos/inmunología , Melanoma , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Macrófagos/patología , Melanoma/inmunología , Melanoma/patología , Melanoma/terapia
8.
Nature ; 475(7355): 222-5, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21654748

RESUMEN

Macrophages, which are abundant in the tumour microenvironment, enhance malignancy. At metastatic sites, a distinct population of metastasis-associated macrophages promotes the extravasation, seeding and persistent growth of tumour cells. Here we define the origin of these macrophages by showing that Gr1-positive inflammatory monocytes are preferentially recruited to pulmonary metastases but not to primary mammary tumours in mice. This process also occurs for human inflammatory monocytes in pulmonary metastases of human breast cancer cells. The recruitment of these inflammatory monocytes, which express CCR2 (the receptor for chemokine CCL2), as well as the subsequent recruitment of metastasis-associated macrophages and their interaction with metastasizing tumour cells, is dependent on CCL2 synthesized by both the tumour and the stroma. Inhibition of CCL2-CCR2 signalling blocks the recruitment of inflammatory monocytes, inhibits metastasis in vivo and prolongs the survival of tumour-bearing mice. Depletion of tumour-cell-derived CCL2 also inhibits metastatic seeding. Inflammatory monocytes promote the extravasation of tumour cells in a process that requires monocyte-derived vascular endothelial growth factor. CCL2 expression and macrophage infiltration are correlated with poor prognosis and metastatic disease in human breast cancer. Our data provide the mechanistic link between these two clinical associations and indicate new therapeutic targets for treating metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Quimiocina CCL2/metabolismo , Inflamación/patología , Monocitos/patología , Metástasis de la Neoplasia , Animales , Antígeno CD11b/metabolismo , Quimiocina CCL2/antagonistas & inhibidores , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Neoplasias Pulmonares/secundario , Macrófagos/patología , Ratones , Monocitos/metabolismo , Metástasis de la Neoplasia/tratamiento farmacológico , Trasplante de Neoplasias , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/metabolismo , Receptores de IgG/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Nat Genet ; 39(4): 467-75, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17369830

RESUMEN

Inactivation of TGF-beta family signaling is implicated in colorectal tumor progression. Using cis-Apc(+/Delta716) Smad4(+/-) mutant mice (referred to as cis-Apc/Smad4), a model of invasive colorectal cancer in which TGF-beta family signaling is blocked, we show here that a new type of immature myeloid cell (iMC) is recruited from the bone marrow to the tumor invasion front. These CD34(+) iMCs express the matrix metalloproteinases MMP9 and MMP2 and the CC-chemokine receptor 1 (CCR1) and migrate toward the CCR1 ligand CCL9. In adenocarcinomas, expression of CCL9 is increased in the tumor epithelium. By deleting Ccr1 in the background of the cis-Apc/Smad4 mutant, we further show that lack of CCR1 prevents accumulation of CD34(+) iMCs at the invasion front and suppresses tumor invasion. These results indicate that loss of transforming growth factor-beta family signaling in tumor epithelium causes accumulation of iMCs that promote tumor invasion.


Asunto(s)
Carcinoma/genética , Movimiento Celular/genética , Neoplasias Intestinales/genética , Células Mieloides/metabolismo , Receptores de Quimiocina/metabolismo , Proteína Smad4/genética , Animales , Antígenos CD34/metabolismo , Carcinoma/patología , Quimiocinas CC , Femenino , Neoplasias Intestinales/patología , Proteínas Inflamatorias de Macrófagos/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Invasividad Neoplásica/genética , Receptores CCR1 , Receptores de Quimiocina/genética , Células del Estroma/metabolismo , Células Tumorales Cultivadas
10.
Pharmacol Res ; 100: 266-70, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26275794

RESUMEN

Metastatic breast cancer is incurable by current therapies including chemotherapy and immunotherapy. Accumulating evidence indicates that tumor-infiltrating macrophages promote establishment of the lethal metastatic foci and contribute to therapeutic resistance. Recent studies suggest that the accumulation of these macrophages is regulated by a chemokine network established in the tumor microenvironment. In this perspective paper, we elaborate on the chemokine signals that can attract monocytes/macrophages to the site of metastasis, and discuss whether inhibition of these chemokine signals can represent a new therapeutic strategy for metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quimiocinas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Microambiente Tumoral/efectos de los fármacos
11.
ACS Cent Sci ; 10(1): 143-154, 2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38292608

RESUMEN

The essential functions that cytokine/immune cell interactions play in tissue homeostasis and during disease have prompted the molecular design of targeted fluorophores to monitor their activity in real time. Whereas activatable probes for imaging immune-related enzymes are common, many immunological functions are mediated by binding events between cytokines and their cognate receptors that are hard to monitor by live-cell imaging. A prime example is interleukin-33 (IL-33), a key cytokine in innate and adaptive immunity, whose interaction with the ST2 cell-surface receptor results in downstream signaling and activation of NF-κB and AP-1 pathways. In the present work, we have designed a chemical platform to site-specifically introduce OFF-to-ON BODIPY fluorophores into full cytokine proteins and generate the first nativelike fluorescent analogues of IL-33. Among different incorporation strategies, chemical aminoacylation followed by bioorthogonal derivatization led to the best labeling results. Importantly, the BODIPY-labeled IL-33 derivatives-unlike IL-33-GFP constructs-exhibited ST2-specific binding and downstream bioactivity profiles comparable to those of the wild-type interleukin. Real-time fluorescence microscopy assays under no wash conditions confirmed the internalization of IL-33 through ST2 receptors and its intracellular trafficking through the endosomal pathway. We envision that the modularity and versatility of our BODIPY labeling platform will facilitate the synthesis of minimally tagged fluorogenic cytokines as the next generation of imaging reagents for real-time visualization of signaling events in live immune cells.

12.
Proc Natl Acad Sci U S A ; 107(29): 13063-8, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20616008

RESUMEN

Recent reports have suggested critical roles of myeloid cells in tumor invasion and metastasis, although these findings have not led to therapeutics. Using a mouse model for liver dissemination, we show that mouse and human colon cancer cells secrete CC-chemokine ligands CCL9 and CCL15, respectively, and recruit CD34(+) Gr-1(-) immature myeloid cells (iMCs). They express CCL9/15 receptor CCR1 and produce matrix metalloproteinases MMP2 and MMP9. Lack of the Ccr1, Mmp2, or Mmp9 gene in the host dramatically suppresses outgrowths of disseminated tumors in the liver. Importantly, CCR1 antagonist BL5923 blocks the iMC accumulation and metastatic colonization and significantly prolongs the survival of tumor-bearing mice. These results suggest that CCR1 antagonists can provide antimetastatic therapies for patients with disseminated colon cancer in the liver.


Asunto(s)
Diferenciación Celular , Neoplasias del Colon/patología , Neoplasias Hepáticas/secundario , Células Mieloides/patología , Receptores CCR1/metabolismo , Animales , Quimiocinas CC/metabolismo , Neoplasias del Colon/metabolismo , Modelos Animales de Enfermedad , Humanos , Ligandos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Proteínas Inflamatorias de Macrófagos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Células Mieloides/metabolismo , Receptores CCR1/antagonistas & inhibidores , Transducción de Señal , Análisis de Supervivencia
13.
Essays Biochem ; 67(6): 1003-1014, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37313600

RESUMEN

Adoptive transfer of natural killer (NK) cells has been proposed as a novel immunotherapy for malignant tumours resistant to current therapeutic modalities. Several clinical studies have demonstrated that the NK cell-infusion is well tolerated without severe side effects and shows promising results in haematological malignancies. However, patients with malignant solid tumours do not show significant responses to this therapy. Such disappointing results largely arise from the inefficient delivery of infused NK cells and the impairment of their functions in the tumour microenvironment (TME). Tumour-associated macrophages (TAMs) are the most abundant stromal cells in the TME of most solid tumours, and a high TAM density correlates with poor prognosis of cancer patients. Although our knowledge of the interactions between TAMs and NK cells is limited, many studies have indicated that TAMs suppress NK cell cytotoxicity against cancer cells. Therefore, blockade of TAM functions can be an attractive strategy to improve NK cell-based immunotherapies. On the other hand, macrophages are reported to activate NK cells under certain circumstances. This essay presents our current knowledge about mechanisms by which macrophages regulate NK cell functions and discusses possible therapeutic approaches to block macrophage-mediated NK cell suppression.


Asunto(s)
Neoplasias , Macrófagos Asociados a Tumores , Humanos , Células Asesinas Naturales , Neoplasias/terapia , Inmunoterapia/métodos , Microambiente Tumoral
14.
PLoS One ; 18(3): e0279057, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36996049

RESUMEN

Although immunotherapy is becoming a standard approach of human cancer treatment, only a small but critical fraction of patients responds to the therapy. It is therefore required to determine the sub-populations of patients who will respond to immunotherapies along with developing novel strategies to improve efficacy of anti-tumor immune reactions. Current development of novel immunotherapies relies heavily on mouse models of cancer. These models are important for better understanding of mechanisms behind tumor immune escape and investigation of novel strategies to overcome it. Nevertheless, the murine models do not necessarily represent the complexity of spontaneously occurring cancers in humans. Dogs spontaneously develop a wide range of cancer types with an intact immune system under similar environment and exposure to humans, which can serve as translational models in cancer immunotherapy research. To date though, there is still a relatively limited amount of information regarding immune cell profiles in canine cancers. One possible reason could be that there are hardly any established methods to isolate and simultaneously detect a range of immune cell types in neoplastic tissues. To date only a single manuscript describes characterization of immune cells in canine tumour tissues, concentrating solely on T-cells. Here we describe a protocol for multi-color flow cytometry to distinguish immune cell types in blood, lymph nodes, and neoplastic tissues from dogs with cancer. Our results demonstrate that a 9-color flow cytometry panel enables characterization of different cell subpopulations including myeloid cells. We also show that the panel allows detection of minor/aberrant subsets within a mixed population of cells in various neoplastic samples including blood, lymph node and solid tumors. To our knowledge, this is the first simultaneous immune cell detection panel applicable for solid tumors in dogs. This multi-color flow cytometry panel has the potential to inform future basic research focusing on immune cell functions in translational canine cancer models.


Asunto(s)
Neoplasias , Animales , Perros , Humanos , Ratones , Citometría de Flujo/veterinaria , Neoplasias/terapia , Linfocitos T , Células Mieloides , Ganglios Linfáticos
15.
Angew Chem Weinheim Bergstr Ger ; 135(8): e202216142, 2023 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38515764

RESUMEN

Cytotoxic immune cells, including T lymphocytes (CTLs) and natural killer (NK) cells, are essential components of the host response against tumors. CTLs and NK cells secrete granzyme A (GzmA) upon recognition of cancer cells; however, there are very few tools that can detect physiological levels of active GzmA with high spatiotemporal resolution. Herein, we report the rational design of the near-infrared fluorogenic substrates for human GzmA and mouse GzmA. These activity-based probes display very high catalytic efficiency and selectivity over other granzymes, as shown in tissue lysates from wild-type and GzmA knock-out mice. Furthermore, we demonstrate that the probes can image how adaptive immune cells respond to antigen-driven recognition of cancer cells in real time.

16.
Angew Chem Weinheim Bergstr Ger ; 134(41): e202207508, 2022 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-38505293

RESUMEN

Increased levels of tumor-associated macrophages (TAMs) are indicators of poor prognosis in most cancers. Although antibodies and small molecules blocking the recruitment of macrophages to tumors are under evaluation as anticancer therapies, these strategies are not specific for macrophage subpopulations. Herein we report the first enzyme-activatable chemokine conjugates for effective targeting of defined macrophage subsets in live tumors. Our constructs exploit the high expression of chemokine receptors (e.g., CCR2) and the activity of cysteine cathepsins in TAMs to target these cells selectively over other macrophages and immune cells (e.g., neutrophils, T cells, B cells). Furthermore, we demonstrate that cathepsin-activatable chemokines are compatible with both fluorescent and therapeutic cargos, opening new avenues in the design of targeted theranostic probes for immune cells in the tumor microenvironment.

17.
Nat Commun ; 13(1): 2366, 2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35501326

RESUMEN

Immunotherapy promotes the attack of cancer cells by the immune system; however, it is difficult to detect early responses before changes in tumor size occur. Here, we report the rational design of a fluorogenic peptide able to detect picomolar concentrations of active granzyme B as a biomarker of immune-mediated anticancer action. Through a series of chemical iterations and molecular dynamics simulations, we synthesize a library of FRET peptides and identify probe H5 with an optimal fit into granzyme B. We demonstrate that probe H5 enables the real-time detection of T cell-mediated anticancer activity in mouse tumors and in tumors from lung cancer patients. Furthermore, we show image-based phenotypic screens, which reveal that the AKT kinase inhibitor AZD5363 shows immune-mediated anticancer activity. The reactivity of probe H5 may enable the monitoring of early responses to anticancer treatments using tissue biopsies.


Asunto(s)
Inmunoterapia , Neoplasias Pulmonares , Animales , Biopsia , Granzimas , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Péptidos , Investigación
18.
Int J Clin Oncol ; 16(5): 464-72, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21847533

RESUMEN

Colorectal cancer is the second most common cancer, and is the third leading cause of cancer-related death in Japan. The majority of these deaths is attributable to liver metastasis. Recent studies have provided increasing evidence that the chemokine-chemokine receptor system is a potential mechanism of tumor metastasis via multiple complementary actions: (a) by promoting cancer cell migration, invasion, survival and angiogenesis; and (b) by recruiting distal stromal cells (i.e., myeloid bone marrow-derived cells) to indirectly facilitate tumor invasion and metastasis. Here, we discuss recent preclinical and clinical data supporting the view that chemokine pathways are potential therapeutic targets for liver metastasis of colorectal cancer.


Asunto(s)
Quimiocinas/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Receptores de Quimiocina/metabolismo , Movimiento Celular/genética , Quimiocinas/antagonistas & inhibidores , Quimiocinas/genética , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Terapia Molecular Dirigida , Células Mieloides/metabolismo , Células Mieloides/patología , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/genética , Microambiente Tumoral
19.
J Immunother Cancer ; 9(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33472858

RESUMEN

BACKGROUND: Metastatic breast cancer is a leading cause of cancer-related death in women worldwide. Infusion of natural killer (NK) cells is an emerging immunotherapy for such malignant tumors, although elimination of the immunosuppressive tumor environment is required to improve its efficacy. The effects of this "metastatic" tumor environment on NK cells, however, remain largely unknown. Previous studies, including our own, have demonstrated that metastasis-associated macrophages (MAMs) are one of the most abundant immune cell types in the metastatic tumor niche in mouse models of metastatic breast cancer. We thus investigated the effects of MAMs on antitumor functions of NK cells in the metastatic tumor microenvironment. METHODS: MAMs were isolated from the tumor-bearing lung of C57BL/6 mice intravenously injected with E0771-LG mouse mammary tumor cells. The effects of MAMs on NK cell cytotoxicity towards E0771-LG cells were evaluated in vitro by real-time fluorescence microscopy. The effects of MAM depletion on NK cell activation, maturation, and accumulation in the metastatic lung were evaluated by flow cytometry (CD69, CD11b, CD27) and in situ hybridization (Ncr1) using colony-stimulating factor 1 (CSF-1) receptor conditional knockout (Csf1r-cKO) mice. Finally, metastatic tumor loads in the chest region of mice were determined by bioluminescence imaging in order to evaluate the effect of MAM depletion on therapeutic efficacy of endogenous and adoptively transferred NK cells in suppressing metastatic tumor growth. RESULTS: MAMs isolated from the metastatic lung suppressed NK cell-induced tumor cell apoptosis in vitro via membrane-bound transforming growth factor ß (TGF-ß) dependent mechanisms. In the tumor-challenged mice, depletion of MAMs increased the percentage of activated (CD69+) and mature (CD11b+CD27-) NK cells and the number of Ncr1+ NK cells as well as NK cell-mediated tumor rejection in the metastatic site. Moreover, MAM depletion or TGF-ß receptor antagonist treatment significantly enhanced the therapeutic efficacy of NK cell infusion in suppressing early metastatic tumor outgrowth. CONCLUSION: This study demonstrates that MAMs are a main negative regulator of NK cell function within the metastatic tumor niche, and MAM targeting is an attractive strategy to improve NK cell-based immunotherapy for metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/terapia , Células Asesinas Naturales/trasplante , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Factor de Crecimiento Transformador beta/metabolismo , Macrófagos Asociados a Tumores/inmunología , Traslado Adoptivo , Animales , Antígenos Ly/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Femenino , Técnicas de Inactivación de Genes , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Trasplante de Neoplasias , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
20.
Angew Chem Weinheim Bergstr Ger ; 133(11): 5763-5767, 2021 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38505495

RESUMEN

Natural killer (NK) cells are immune cells that can kill certain types of cancer cells. Adoptive transfer of NK cells represents a promising immunotherapy for malignant tumours; however, there is a lack of methods to validate anti-tumour activity of NK cells in vivo. Herein, we report a new chemiluminescent probe to image in situ the granzyme B-mediated killing activity of NK cells against cancer cells. We have optimised a granzyme B-specific construct using an activatable phenoxydioxetane reporter so that enzymatic cleavage of the probe results in bright chemiluminescence. The probe shows high selectivity for active granzyme B over other proteases and higher signal-to-noise ratios than commercial fluorophores. Finally, we demonstrate that the probe can detect NK cell activity in mouse models, being the first chemiluminescent probe for in vivo imaging of NK cell activity in live tumours.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA