Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 31(7): 2105-2119, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37073129

RESUMEN

Poor intratumoral infiltration is the major challenge for chimeric antigen receptor (CAR)-T cell therapy in solid tumors. Hypofractionated radiotherapy (HFRT) has been reported to induce immune cell infiltration and reshape the tumor immune microenvironment. Here, we showed that HFRT (5 × 5 Gy) mediated an early accumulation of intratumoral myeloid-derived suppressor cells (MDSCs) and decreased infiltration of T cells in the tumor microenvironment (TME) of immunocompetent mice bearing triple-negative breast cancer (TNBC) or colon cancer, which was further confirmed in tumors from patients. RNA sequencing (RNA-seq) and cytokine profiling analysis revealed that HFRT induced the activation and proliferation of tumor-infiltrated MDSCs, which was mediated by the interactions of multiple chemokines and chemokine receptors. Further investigation showed that when combined with HFRT, CXCR2 blockade significantly inhibited MDSCs trafficking to tumors and effectively enhanced the intratumoral infiltration and treatment efficacy of CAR-T cells. Our study demonstrates that MDSCs blockade combined with HFRT is promising for CAR-T cell therapy optimization in solid tumors.


Asunto(s)
Células Supresoras de Origen Mieloide , Receptores Quiméricos de Antígenos , Ratones , Animales , Receptores Quiméricos de Antígenos/genética , Línea Celular Tumoral , Inmunoterapia Adoptiva , Linfocitos T , Microambiente Tumoral
2.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(2): 145-151, 2019 Mar.
Artículo en Zh | MEDLINE | ID: mdl-31106530

RESUMEN

OBJECTIVE: To test the killing effect of type Ⅰ receptor tyrosine kinase-like orphan receptor (ROR1) chimeric antigen receptor T cell (CAR-T) on several ROR1-expressing tumor cells in vitro. METHODS: The CAR gene was designed and synthesized by constructing the lentiviral vector plasmid, and BamHⅠ/EcoRⅠ was used to identify the plasmid. The expression levels of ROR1 among a variety of tumor cell lines were compared using flow cytometry (FCM). The killing effect of CAR-T on positive cells was detected by FCM, the LDH assay and ELISA. RESULTS: The double enzyme digestion identified CAR gene was successfully constructed to the lentivirus vector plasmid. FCM detection showed that the efficiency of CAR-T infection was about 47.23%. Multiple tumor cells expressed ROR1 in varying degrees. The FCM and the LDH assay indicated that CAR-T specifically killed ROR1-positive tumor cells. On positive target cells, more interferonI-γ (FN-γ) could be released during the CAR-T killing process than control T (P<0.05). CONCLUSION: We successfully constructed ROR1 CAR-T. CAR-T can specifically kill ROR1-positive tumor cells and cause the release of large amounts of IFN-γ, providing an experimental basis for clinical application.


Asunto(s)
Inmunoterapia Adoptiva , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/inmunología , Receptores de Antígenos de Linfocitos T , Receptores Quiméricos de Antígenos , Linfocitos T/citología , Línea Celular Tumoral , Humanos , Lentivirus
3.
Anticancer Drugs ; 27(8): 711-22, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27333595

RESUMEN

T-lymphocytes genetically engineered with the chimeric antigen receptor (CAR-T) have shown great therapeutic potential in cancer treatment. A variety of preclinical researches and clinical trials of CAR-T therapy have been carried out to lay the foundation for future clinical application. In these researches, several gene-transfer methods were used to deliver CARs or other genes into T-lymphocytes, equipping CAR-modified T cells with a property of recognizing and attacking antigen-expressing tumor cells in a major histocompatibility complex-independent manner. Here, we summarize the gene-transfer vectors commonly used in the generation of CAR-T cell, including retrovirus vectors, lentivirus vectors, the transposon/transposase system, the plasmid-based system, and the messenger RNA electroporation system. The following aspects were compared in parallel: efficiency of gene transfer, the integration methods in the modified T cells, foreground of scale-up production, and application and development in clinical trials. These aspects should be taken into account to generate the optimal CAR-gene vector that may be suitable for future clinical application.


Asunto(s)
Vectores Genéticos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/fisiología , Transducción Genética/métodos , Transfección/métodos , Animales , Ensayos Clínicos como Asunto , Elementos Transponibles de ADN , Electroporación/métodos , Humanos , Inmunoterapia/métodos , Receptores de Antígenos de Linfocitos T/metabolismo
4.
J Gastroenterol Hepatol ; 29(3): 463-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24117871

RESUMEN

BACKGROUND AND AIM: A strong association between family function and irritable bowel syndrome (IBS) has been observed. Parental rearing styles, as a comprehensive mark for family function, may provide new clues to the etiology of IBS. This study aimed to explore which dimensions of parental rearing styles were risk factors or protective factors for IBS in adolescents. METHODS: Two thousand three hundred twenty adolescents were recruited from one middle school and one high school randomly selected from Jiangan District (an urban district in Wuhan City). Data were collected using two Chinese versions of validated self-report questionnaires including the Rome III diagnostic criteria for pediatric IBS and the Egna Minnen Beträffande Uppfostran: One's Memories of Upbringing for perceived parental rearing styles. RESULTS: Ninety-six subjects diagnosed as pediatric IBS were compared with 1618 controls. The IBS patients reported less both paternal and maternal emotional warmth (all P < 0.01) and more both paternal and maternal punishment, overinterference, rejection, and overprotection (only for father) (all P < 0.01) than the controls. Furthermore, the IBS patients had higher total scores of parental rearing styles (all P < 0.001) than the controls. With univariate logistic regression, standardized regression coefficients and odds ratios of parental rearing variables were calculated. Multivariate logistic regression revealed that paternal rejection (P = 0.001) and maternal overinterference (P = 0.002) were independent risk factors for IBS in adolescents. CONCLUSIONS: Parental emotional warmth is a protective factor for IBS in adolescents and parental punishment, overinterference, rejection, and overprotection are risk factors for IBS in adolescents.


Asunto(s)
Crianza del Niño/psicología , Síndrome del Colon Irritable/etiología , Síndrome del Colon Irritable/psicología , Relaciones Padres-Hijo , Padres/psicología , Adolescente , Niño , Emociones/fisiología , Femenino , Humanos , Síndrome del Colon Irritable/epidemiología , Síndrome del Colon Irritable/prevención & control , Modelos Logísticos , Masculino , Castigo/psicología , Rechazo en Psicología , Factores de Riesgo , Encuestas y Cuestionarios
5.
Front Immunol ; 15: 1325946, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38500876

RESUMEN

Immunotherapies have revolutionized the landscape of cancer treatment. Regulatory T cells (Tregs), as crucial components of the tumor immune environment, has great therapeutic potential. However, nonspecific inhibition of Tregs in therapies may not lead to enhanced antitumor responses, but could also trigger autoimmune reactions in patients, resulting in intolerable treatment side effects. Hence, the precision targeting and inhibition of tumor-infiltrating Tregs is of paramount importance. In this overview, we summarize the characteristics and subpopulations of Tregs within tumor microenvironment and their inhibitory mechanisms in antitumor responses. Furthermore, we discuss the current major strategies targeting regulatory T cells, weighing their advantages and limitations, and summarize representative clinical trials targeting Tregs in cancer treatment. We believe that developing therapies that specifically target and suppress tumor-infiltrating Tregs holds great promise for advancing immune-based therapies.


Asunto(s)
Neoplasias , Humanos , Inmunoterapia/métodos , Linfocitos T Reguladores , Microambiente Tumoral
6.
Curr Med Chem ; 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38310398

RESUMEN

Cancer immunotherapy has demonstrated remarkable success in the treatment of multiple advanced malignancies, especially approaches to target the immune checkpoint. Nonetheless, the limited response rate remains a barrier to broader application. Identifying other ways to extend the beneficiaries to a large extent is needed. Emerging evidence has shown that mitogen-activated protein kinase-interacting kinases (MNKs) could be regarded as a novel, attractive target for cancer immunotherapy that is closely correlated with cancer biology and therapies. A comprehensive understanding of the role and mechanism of MNKs in cancer will shed light on the discovery of novel therapeutic strategies for cancer treatment. In this review, we outlined the structure of MNKs, their function and expression, and how MNKs affect tumor progression and elucidated the evidence supporting MNKs as a new promising treatment modality in human cancers.

7.
Front Immunol ; 14: 1095943, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36776869

RESUMEN

Although breakthroughs in cancer treatment have been achieved, immunotherapy yields only modest benefits in most patients. There is still a gap in clarifying the immune evasiveness and immune-resistance mechanisms. Identifying other candidate targets for cancer immunotherapy is therefore a clear unmet clinical need. The complement system, a pillar of innate immunity, has recently entered the limelight due to its immunoregulatory functions in the tumor microenvironment (TME). In particular, gC1qR, a receptor for globular heads of C1q, serves as a promising new target and has attracted more attention. gC1qR, also named P32/C1qBP/HABP1, is a multifunctional protein that is overexpressed in various cancers and holds prognostic value. It regulates the tumorigenic, progression and metastatic properties of tumor cells through several downstream signaling pathways, including the Wnt/ß-catenin, PKC-NF-κB and Akt/PKB pathways. A few preclinical experiments conducted through gC1qR interventions, such as monoclonal antibody, chimeric antigen receptor T-cell (CAR-T) therapy, and tumor vaccination, have shown encouraging results in anticancer activity. The efficacy may rely on the regulatory role on the TME, induction of tumor cells apoptosis and antiangiogenic activity. Nevertheless, the current understanding of the relationship between cancer immunotherapy and gC1qR remains elusive and often contradictory, posing both opportunities and challenges for therapeutic translation in the clinic. In this review, we focus on the current understanding of gC1qR function in cancer immunology and highlight the vital roles in regulating the TME. We also examines the rationale behind targeting gC1qR and discusses the potential for translating into clinical practice.


Asunto(s)
Proteínas Portadoras , Neoplasias , Humanos , Proteínas Portadoras/metabolismo , Neoplasias/terapia , Transducción de Señal , Inmunoterapia , Microambiente Tumoral , Proteínas Mitocondriales/metabolismo
8.
J Immunother Cancer ; 11(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37591632

RESUMEN

BACKGROUNDS: Chimeric antigen receptor (CAR)-modified T cells (CAR-T) are limited in solid tumors due to the hostile tumor microenvironment (TME). Combination therapy could be a promising approach to overcome this obstacle. Recent studies have shown that sphingosine 1-phosphate receptor (S1PR)3 has tremendous potential in regulating the immune environment. However, the functional significance of S1PR3 in T-cell-based immunotherapies and the molecular mechanisms have not been fully understood. METHODS: Here, we studied the combination of EpCAM-specific CAR T-cell therapy with pharmacological blockade of S1PR3 against solid tumor. We have applied RNA sequencing, flow cytometry, ELISA, cellular/molecular immunological technology, and mouse models of solid cancers. RESULTS: Our study provided evidence that S1PR3 high expression is positively associated with resistance to programmed cell death protein-1 (PD-1)-based immunotherapy and increased T-cell exhaustion. In addition, pharmacological inhibition of S1PR3 improves the efficacy of anti-PD-1 therapy. Next, we explored the possible combination of S1PR3 antagonist with murine EpCAM-targeted CAR-T cells in immunocompetent mouse models of breast cancer and colon cancer. The results indicated that the S1PR3 antagonist could significantly enhance the efficacy of murine EpCAM CAR-T cells in vitro and in vivo. Mechanistically, the S1PR3 antagonist improved CAR-T cell activation, regulated the central memory phenotype, and reduced CAR-T cell exhaustion in vitro. Targeting S1PR3 was shown to remodel the TME through the recruitment of proinflammatory macrophages by promoting macrophage activation and proinflammatory phenotype polarization, resulting in improved CAR-T cell infiltration and amplified recruitment of CD8+T cells. CONCLUSIONS: This work demonstrated targeting S1PR3 could increase the antitumor activities of CAR-T cell therapy at least partially by inhibiting T-cell exhaustion and remodeling the TME through the recruitment of proinflammatory macrophages. These findings provided additional rationale for combining S1PR3 inhibitor with CAR-T cells for the treatment of solid tumor.


Asunto(s)
Neoplasias del Colon , Receptores Quiméricos de Antígenos , Animales , Ratones , Molécula de Adhesión Celular Epitelial , Receptores de Esfingosina-1-Fosfato , Agotamiento de Células T , Modelos Animales de Enfermedad , Microambiente Tumoral
9.
J Clin Med ; 12(19)2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-37834768

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of malignancies, especially hematological tumors, but toxicities have tempered its success. The main impediments to the development of CAR-T cell therapies are the following: cytokine release syndrome (CRS), immune-effector-cell-associated neurotoxicity syndrome (ICANS), tumor lysis syndrome (TLS), and on-target/off-tumor toxicity (OTOT). This review summarizes these side effects' underlying mechanisms and manifestations over time. It provides potential prevention and treatment according to the consensus grading, stressing the significance of establishing strategies that anticipate, reduce, and navigate the beginning of these side effects. It is essential to fully comprehend the mechanisms underlying these toxicities to create efficient treatment and preventive approaches.

10.
J Oncol ; 2022: 4259205, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35087588

RESUMEN

Tumor immunotherapy brings substantial and long-term clinical benefits that can even cure tumors. However, the accumulation of evidence suggests that immunotherapy also induces severe and complex neurologic immune-related adverse events (ir-AEs) and even leads to immunotherapy-related death, which arouses the concern of clinicians. The timely and accurate identification of neurotoxicity helps clinicians detect and treat these complications early, thereby enhancing treatment efficiency and improving the prognosis of patients. At present, the mechanism of neurotoxicity caused by immunotherapy has not been completely elucidated. This paper mainly reviews the clinical features, pathogenesis, and therapeutic strategies of neurologic ir-AEs.

11.
Genome Biol ; 23(1): 269, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36575517

RESUMEN

Most single-cell RNA sequencing (scRNA-seq) analyses begin with cell clustering; thus, the clustering accuracy considerably impacts the validity of downstream analyses. In contrast with the abundance of clustering methods, the tools to assess the clustering accuracy are limited. We propose a new Clustering Deviation Index (CDI) that measures the deviation of any clustering label set from the observed single-cell data. We conduct in silico and experimental scRNA-seq studies to show that CDI can select the optimal clustering label set. As a result, CDI also informs the optimal tuning parameters for any given clustering method and the correct number of cluster components.


Asunto(s)
Algoritmos , Perfilación de la Expresión Génica , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Análisis de Expresión Génica de una Sola Célula , Análisis de la Célula Individual/métodos , Análisis por Conglomerados
12.
Nat Cell Biol ; 24(2): 230-241, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35145222

RESUMEN

Many cancers have an unusual dependence on glutamine. However, most previous studies have focused on the contribution of glutamine to metabolic building blocks and the energy supply. Here, we report that cancer cells with aberrant expression of glutamate decarboxylase 1 (GAD1) rewire glutamine metabolism for the synthesis of γ-aminobutyric acid (GABA)-a prominent neurotransmitter-in non-nervous tissues. An analysis of clinical samples reveals that increased GABA levels predict poor prognosis. Mechanistically, we identify a cancer-intrinsic pathway through which GABA activates the GABAB receptor to inhibit GSK-3ß activity, leading to enhanced ß-catenin signalling. This GABA-mediated ß-catenin activation both stimulates tumour cell proliferation and suppresses CD8+ T cell intratumoural infiltration, such that targeting GAD1 or GABABR in mouse models overcomes resistance to anti-PD-1 immune checkpoint blockade therapy. Our findings uncover a signalling role for tumour-derived GABA beyond its classic function as a neurotransmitter that can be targeted pharmacologically to reverse immunosuppression.


Asunto(s)
Proliferación Celular , Neoplasias/metabolismo , Escape del Tumor , Microambiente Tumoral/inmunología , beta Catenina/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Células A549 , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Receptores de GABA-B/metabolismo , Carga Tumoral , Escape del Tumor/efectos de los fármacos , Vía de Señalización Wnt , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
13.
Signal Transduct Target Ther ; 7(1): 380, 2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36402752

RESUMEN

Pleural and peritoneal metastasis accompanied by malignant pleural effusion (MPE) or malignant ascites (MA) is frequent in patients with advanced solid tumors that originate from the lung, breast, gastrointestinal tract and ovary. Regional delivery of CAR-T cells represents a new strategy to control tumor dissemination in serous cavities. However, malignant effusions constitute an immune-suppressive environment that potentially induces CAR-T cell dysfunction. Here, we demonstrated that the anti-tumor cytotoxicity of conventional 2nd-generation CAR-T cells was significantly inhibited by both the cellular and non-cellular components of MPE/MA, which was primarily attributed to impaired CAR-T cell proliferation and cytokine production in MPE/MA environment. Interestingly, we found that PD-L1 was widely expressed on freshly-isolated MPE/MA cells. Based on this feature, a novel PD-L1-targeting chimeric switch receptor (PD-L1.BB CSR) was designed, which can bind to PD-L1, switching the inhibitory signal into an additional 4-1BB signal. When co-expressed with a 2nd-generation CAR, PD-L1.BB CSR-modified CAR-T cells displayed superior fitness and enhanced functions in both culture medium and MPE/MA environment, causing rapid and durable eradication of pleural and peritoneal metastatic tumors in xenograft models. Further investigations revealed elevated expressions of T-cell activation, proliferation, and cytotoxicity-related genes, and we confirmed that PD-L1 scFv and 4-1BB intracellular domain, the two important components of PD-L1.BB CSR, were both necessary for the functional improvements of CAR-T cells. Overall, our study shed light on the clinical application of PD-L1.BB CSR-modified dual-targeting CAR-T cells. Based on this study, a phase I clinical trial was initiated in patients with pleural or peritoneal metastasis (NCT04684459).


Asunto(s)
Neoplasias Peritoneales , Derrame Pleural Maligno , Femenino , Humanos , Antígeno B7-H1/genética , Activación de Linfocitos , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/terapia , Derrame Pleural Maligno/metabolismo , Linfocitos T , Ensayos Clínicos Fase I como Asunto
14.
Theranostics ; 11(10): 4699-4709, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754022

RESUMEN

Rationale: The onset of cytokine release syndrome (CRS) and in vivo persistence of anti-CD19 chimeric antigen receptor T (CAR-T) cells after infusion correlate with clinical responsiveness. However, there are no known baseline biomarkers that can predict the prognosis of patients with B-lineage non-Hodgkin lymphoma (B-NHL). The aim of this study was to identify blood cell populations associated with beneficial outcomes in B-NHL patients administered CAR-T cell immunotherapies. Methods: We enumerated peripheral blood and CAR-T cells by retrospectively analyzing three CAR-T cell trials involving 65 B-NHL patients. We used a preclinical model to elucidate the eosinophil mechanism in CAR-T cell therapy. Results: During an observation period up to 30 mo, B-NHL patients with higher baseline eosinophil counts had higher objective response rates than those with low eosinophil counts. Higher baseline eosinophil counts were also significantly associated with durable progression-free survival (PFS). The predictive significance of baseline eosinophil counts was validated in two independent cohorts. A preclinical model showed that eosinophil depletion impairs the intratumoral infiltration of transferred CAR-T cells and reduces CAR-T cell antitumor efficacy. Conclusion: The results of this study suggest that peripheral eosinophils could serve as stratification biomarkers and a recruitment machinery to facilitate anti-CD19 CAR-T cell therapy in B-NHL patients.


Asunto(s)
Eosinófilos , Inmunoterapia Adoptiva , Linfoma de Células B/terapia , Receptores Quiméricos de Antígenos , Adulto , Anciano , Animales , Antígenos CD19 , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Recuento de Leucocitos , Linfoma de Células B/sangre , Masculino , Ratones , Persona de Mediana Edad , Pronóstico , Supervivencia sin Progresión , Adulto Joven
15.
Oncoimmunology ; 9(1): 1806009, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32923168

RESUMEN

The tumoricidal efficiency of human CAR-T cells is generally evaluated using immune-deficient mouse models; however, due to their immune-incompetency and the species-specific reactivity of a target antigen, these models are problematic to imitate CAR-T-induced adverse effects in the clinic. Epithelial cell adhesion molecule (EpCAM) is a tumor-associated antigen overtly presented on the cell surface of various carcinomas, making it an attractive target for CAR-T therapy. Here, we developed an anti-mouse EpCAM CAR to evaluate its safety and efficacy in immunocompetent mouse models. As previously reported for their human equivalents, murine EpCAM CAR-T cells exhibit promising anti-tumor efficacy in vitro and in vivo. However, after CAR-T infusion, various dose-depended toxicities including body weight loss, cytokine-release syndrome (CRS), and death were observed in both tumor-bearing and tumor-free mice. Pathological examination revealed unexpected and severe pulmonary immunopathology due to basal EpCAM expression in normal lung. While our study validates EpCAM CAR-T's potent anti-tumor efficacy, it also reveals that EpCAM CAR-T cells used for the treatment of solid tumors may cause lethal toxicity and should, therefore, be evaluated in patients with caution.


Asunto(s)
Receptores Quiméricos de Antígenos , Animales , Molécula de Adhesión Celular Epitelial , Humanos , Inmunoterapia Adoptiva , Pulmón , Ratones , Linfocitos T
16.
Int J Radiat Oncol Biol Phys ; 107(4): 804-814, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32334032

RESUMEN

PURPOSE: To determine the role of NLRP3 inflammasome activation in low-dose radiation-induced radiation pneumonitis (RP) and to assess whether inhibition or deletion of the NLRP3 inflammasome is critical for conferring protection against RP. METHODS AND MATERIALS: The human monocytic THP-1 cells were treated with increasing doses of radiation to assess the activation of NLRP3 by Western blot and enzyme-linked immunosorbent assay. Reactive oxygen species (ROS) production was measured by flow cytometry, with or without ROS inhibitor treatment. A mouse thoracic radiation model that received different doses of radiation was used, and the lung tissues of thoracic-irradiated nlrp3-/- and wild-type C57BL/6 mice were examined by hematoxylin and eosin and immunofluorescence staining. The concentrations of cytokines in the bronchoalveolar lavage fluid were measured by enzyme-linked immunosorbent assay and Luminex multiplex assays. Lipopolysaccharide (LPS) was administered intranasally 28 days after thoracic irradiation, and NLRP3 inhibitor, MCC950 was administered intraperitoneally after irradiation at 2 different doses. RESULTS: (1) The NLRP3 inflammasome was activated in 2 Gy irradiated THP-1 cells; NLRP3 and cleaved-caspase-1 levels were not associated with dose escalation of irradiation. (2) Activation of the NLRP3 inflammasome was mediated by ROS, and ROS inhibitor treatment decreased the production of IL-1ß and IL-18 in vitro. (3) NLRP3 was activated in mouse lungs by irradiation at 2 Gy, 4 Gy, and 16 Gy, and NLRP3 activation was continuous for 8 weeks. (4) NLRP3 deletion protects against LPS-mediated monocyte infiltration in the mouse lung. (5) The administration of MCC950 decreased the inflammation score of the mice irradiated with 2 Gy or 16 Gy in vivo. CONCLUSIONS: Our results indicate that the NLRP3 inflammasome is activated by low-dose irradiation both in vitro and in vivo. Inhibition or deletion of NLRP3 can specifically alleviate the mouse lung inflammation caused by radiation and LPS treatment. This study reveals the mechanism of low-dose radiation therapy-induced RP and offers a possible treatment strategy.


Asunto(s)
Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Dosis de Radiación , Neumonitis por Radiación/etiología , Neumonitis por Radiación/metabolismo , Línea Celular Tumoral , Humanos , Inflamasomas/antagonistas & inhibidores , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/efectos de la radiación , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Neumonitis por Radiación/inmunología , Neumonitis por Radiación/prevención & control , Especies Reactivas de Oxígeno/metabolismo
17.
Precis Clin Med ; 2(3): 197-203, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35694436

RESUMEN

We report the case of a 90-year-old female patient who was suffering from c-ros oncogene 1 (ros-1) rearrangement adenocarcinoma and breast cancer. After about 14 months of a reduced dose of crizotinib treatment, she had a stable disease according to the Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1). This patient's case demonstrates that ros-1 rearrangements are not limited to patients of young age. In addition, this case indicates that crizotinib, as second-line, or even first-line, treatment may be effective and manageable in elderly patients. Furthermore, for elderly patients carrying a ros1 fusion, a reduced dose of crizotinib may be efficacious rather than a resistance factor. Based on our findings, we recommend that elderly patients with advanced lung adenocarcinoma should be considered for inclusion in molecular screening for ros-1 translocation, especially for never-smokers negative for epidermal growth factor receptor (egfr) mutation and the fusion between echinoderm microtubule associated protein-like 4 (EML4) and anaplastic lymphoma kinase (ALK). This deserves attention because the population is aging, with increasing incidence and morbidity of multiple primary malignant tumors. Neglect of breast nodules at the onset is one of the limitations of our case, as combination of primary lung cancer with breast cancer is common. Above all, use of antiestrogens before and after the diagnosis of non-small-cell lung cancer is related to a reduced risk of lung cancer mortality. Therefore, careful attention should always be paid to these cases.

18.
Hum Gene Ther ; 30(4): 402-412, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30693795

RESUMEN

Chimeric antigen receptor-modified T cells (CAR-T cells) have emerged as a promising cancer immunotherapy for solid tumors. Epithelial cell adhesion molecule (EpCAM) is overexpressed in a variety of tumors and is recognized as a biomarker for circulating tumor cells and cancer stem cells, representing an attractive target for adoptive T-cell immunotherapy. This study generated third-generation CAR-T cells with redirected specificity to EpCAM (EpCAM CAR-T) by lentiviral vector. The study demonstrated that EpCAM CAR-T cells can elicit lytic cytotoxicity to target cells in an EpCAM-dependent manner and secrete cytotoxic cytokines, including interferon gamma and tumor necrosis factor alpha. Furthermore, adoptive transfer of EpCAM CAR-T cells significantly delayed tumor growth and formation in xenograft models. In addition, the safety evaluation showed that CAR-T cells have no systemic toxicity in mice. The data confirmed the antitumor ability and safety of CAR-T cells targeting EpCAM and may provide a new target for CAR-T cell therapies in treating solid tumors.


Asunto(s)
Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Molécula de Adhesión Celular Epitelial/inmunología , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Animales , Biomarcadores , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Molécula de Adhesión Celular Epitelial/antagonistas & inhibidores , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Nat Med ; 24(10): 1536-1544, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30297899

RESUMEN

Impaired immunity in patients with late-stage cancer is not limited to antitumor responses, as demonstrated by poor vaccination protection and high susceptibility to infection1-3. This has been largely attributed to chemotherapy-induced impairment of innate immunity, such as neutropenia2, whereas systemic effects of tumors on hematopoiesis and adoptive immunity remain incompletely understood. Here we observed anemia associated with severe deficiency of CD8+ T cell responses against pathogens in treatment-naive mice bearing large tumors. Specifically, we identify CD45+ erythroid progenitor cells (CD71+TER119+; EPCs) as robust immunosuppressors. CD45+ EPCs, induced by tumor growth-associated extramedullary hematopoiesis, accumulate in the spleen to become a major population, outnumbering regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). The CD45+ EPC transcriptome closely resembles that of MDSCs, and, like MDSCs, reactive oxygen species production is a major mechanism underlying CD45+ EPC-mediated immunosuppression. Similarly, an immunosuppressive CD45+ EPC population was detected in patients with cancer who have anemia. These findings identify a major population of immunosuppressive cells that likely contributes to the impaired T cell responses commonly observed in patients with advanced cancer.


Asunto(s)
Anemia/inmunología , Células Precursoras Eritroides/inmunología , Células Supresoras de Origen Mieloide/inmunología , Sarcoma Mieloide/inmunología , Anemia/genética , Anemia/patología , Animales , Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Humanos , Tolerancia Inmunológica , Inmunidad Innata/genética , Antígenos Comunes de Leucocito/inmunología , Ratones , Estadificación de Neoplasias , Especies Reactivas de Oxígeno/metabolismo , Receptores de Transferrina/inmunología , Sarcoma Mieloide/metabolismo , Sarcoma Mieloide/patología , Linfocitos T Reguladores/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Sci China Life Sci ; 59(4): 333-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26965523

RESUMEN

T cells, genetically modified by chimeric antigen receptors (CAR-T), are endowed with specificity to a desired antigen and are cytotoxic to cells expressing the targeted antigen. CAR-T-based cancer immunotherapy is a promising therapy for curing hematological malignancy, such as acute lymphoid leukemia, and is promising for extending their efficacy to defeat solid tumors. To date, dozens of different CAR-T cells have been evaluated in clinical trials to treat tumors; this necessitates the establishment of guidelines for the production and application of CAR-T cells. However, it is challenging to standardize CAR-T cancer therapy because it involves a combination of gene therapy and cell therapy. In this review, we compare the existing guidelines for CAR-T cells and discuss the challenges and considerations for establishing guidance for CAR-T-based cancer immunotherapy.


Asunto(s)
Guías como Asunto , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/trasplante , Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/normas , Citometría de Flujo , Humanos , Inmunoterapia Adoptiva/normas , Neoplasias/genética , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transfección/métodos , Transfección/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA