Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Transl Med ; 21(1): 321, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37173692

RESUMEN

BACKGROUND: The ubiquitin protein ligase E3C (UBE3C) has been reported to play an oncogenic role in breast cancer (BRCA). This work further investigates the effect of UBE3C on the radioresistance of BRCA cells. METHODS: Molecules linking to radioresistance in BRCA were identified by analyzing two GEO datasets, GSE31863 and GSE101920. UBE3C overexpression or knockdown was induced in parental or radioresistant BRCA cells, followed by irradiation treatment. The malignant properties of cells in vitro, and the growth and metastatic activity of cells in nude mice, were analyzed. Downstream target proteins, as well as upstream transcriptional regulators of UBE3C, were predicted by bioinformatics tools. Molecular interactions were confirmed by immunoprecipitation and immunofluorescence assays. Furthermore, artificial alterations of TP73 and FOSB were induced in the BRCA cells for functional rescue assays. RESULTS: According to bioinformatics analyses, UBE3C expression was linked to radioresistance in BRCA. UBE3C knockdown in radioresistant BRCA cells reduced while its overexpression in parental BRCA cells increased the radioresistance of cells in vitro and in vivo. UBE3C, which induced ubiquitination-dependent protein degradation of TP73, was transcriptionally activated by FOSB. The radioresistance of cancer cells was blocked by TP73 overexpression or FOSB knockdown. Additionally, LINC00963 was found to be responsible for the recruitment of FOSB to the UBE3C promoter for transcription activation. CONCLUSION: This work demonstrates that LINC00963 induces nuclear translocation of FOSB and the consequent transcription activation of UBE3C, which enhances radioresistance of BRCA cells by inducing ubiquitination-dependent protein degradation of TP73.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas c-fos , ARN Largo no Codificante , Tolerancia a Radiación , Ubiquitina-Proteína Ligasas , Animales , Ratones , Línea Celular Tumoral , Ratones Desnudos , Neoplasias/genética , Neoplasias/radioterapia , Proteolisis , Proteínas Proto-Oncogénicas c-fos/genética , Activación Transcripcional/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , ARN Largo no Codificante/genética
2.
Neoplasma ; 69(4): 807-819, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35471977

RESUMEN

This article reported the mechanism of Anlotinib in gastric cancer treatment. Gastric cancer cells were treated with Anlotinib (8 µM) and transfected by STING shRNA and STING vectors. Cell counting kit-8 assay, wounding healing assay, and Transwell experiment were applied for proliferation, migration, and invasion detection. PD-L1 fluorescence intensity in gastric cancer cells was explored by flow cytometry. IFN-ß level was researched by enzyme-linked immunosorbent reaction. Xenograft tumor experiment was performed by administering mice with Anlotinib and anti-PD-L1 antibody. Immunohistochemistry and western blot were used for proteins expression detection. Quantitative real-time reverse transcription-polymerase chain reaction was applied for mRNA expression detection. Hematoxylin and eosin staining was conducted on lung, liver, kidney, and cerebral cortex of mice. Gastric cancer cells treated with Anlotinib exhibited reduced proliferation, migration, and invasion (p<0.01). Anlotinib treatment reduced PCNA, CDK1, and MMP2 protein expressions and increased E-cadherin protein expression in gastric cancer cells (p<0.01). Anlotinib treatment suppressed PD-L1 expression and activated the cGAS-STING/IFN-ß pathway in gastric cancer cells (p<0.01). STING knockdown partially reversed the inhibition of Anlotinib on gastric cancer cells proliferation, migration, invasion, and immune escape (p<0.05 or p<0.01). However, STING overexpression exhibited the opposite effect. Anlotinib synergistically improved anti-tumor efficacy of anti-PD-L1 in vivo. Anlotinib synergistic anti-PD-L1 increased CD3+, CD8+ T cells, and activated the cGAS-STING/IFN-ß pathway in xenograft tumor. Anlotinib was non-toxic to lung, liver, cortex, and kidney. Anlotinib suppressed gastric cancer cells proliferation, migration, and immune escape by activating the cGAS-STING/IFN-ß pathway.


Asunto(s)
Quinolinas , Neoplasias Gástricas , Animales , Proliferación Celular , Humanos , Indoles , Ratones , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Quinolinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico
3.
J Cell Mol Med ; 24(7): 4286-4297, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32100392

RESUMEN

Oncolytic Newcastle disease virus (NDV) induces immunogenic cell death (ICD), liberating danger-associated molecular patterns (DAMPs) that provokes defiance in neoplastic malignancy. The present study aims to investigate whether and how oncolytic NDV triggers ICD in prostate cancer cells. We show that NDV/FMW, an oncolytic NDV strain FMW, elicited the expression and release of several ICD markers, that is calreticulin (CRT), heat shock proteins (HSP70/90) and high-mobility group box 1 (HMGB1), in prostate cancer cells. Furthermore, pharmacological repression of apoptosis, necroptosis, autophagy or endoplasmic reticulum (ER) stress exerted diverse effects on the HMGB1 and HSP70/90 evacuation in NDV/FMW-infected prostate cancer cells. Moreover, ICD markers induced in prostate cancer cells upon NDV/FMW infection, were enhanced by either treatment with a STAT3 (signal transducer and activator of transcription 3) inhibitor or shRNA-mediated knockdown of STAT3. In nude mice bearing prostate cancer cell-derived tumours, the tumours injected with the supernatants of NDV/FMW-infected cells grew smaller than mock-treated tumours. These results indicate that oncolytic NDV provokes the expression of ICD makers in prostate cancer cells. Our data also suggest that a combination of inhibition of STAT3 with oncolytic NDV could boost NDV-based anti-tumour effects against prostate cancer.


Asunto(s)
Muerte Celular Inmunogénica/genética , Viroterapia Oncolítica , Neoplasias de la Próstata/genética , Factor de Transcripción STAT3/genética , Animales , Apoptosis/genética , Autofagia/genética , Calreticulina/genética , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteína HMGB1/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/genética , Humanos , Masculino , Ratones , Ratones Desnudos , Necroptosis/genética , Virus de la Enfermedad de Newcastle/genética , Virus Oncolíticos/genética , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/virología , Factor de Transcripción STAT3/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Oncologist ; 25(11): e1671-e1680, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32627883

RESUMEN

BACKGROUND: The increasing molecular characterization of colorectal cancers (CRCs) has spurred the need to look beyond RAS, BRAF, and microsatellite instability (MSI). Genomic alterations, including ERBB2 amplifications and mutations, POLE mutations, MSI, and NTRK1-3 fusions, have emerged as targets for matched therapies. We sought to study a clinically annotated Chinese cohort of CRC subjected to genomic profiling to explore relative target frequencies. METHODS: Tumor and matched whole blood were collected from 609 Chinese patients with CRC. Extracted DNA was analyzed for all classes of genomic alterations across 450 cancer-related genes, including single-nucleotide variations (SNVs), short and long insertions and deletions (indels), copy number variations, and gene rearrangements. Next-generation sequencing-based computational algorithms also determined tumor mutational burden and MSI status. RESULTS: Alterations in TP53 (76%), APC (72%), and KRAS (46%) were common in Chinese patients with CRC. For the first time, the prevalence of NTRK gene fusion was observed to be around 7% in the MSI-high CRC cohort. Across the cohort, MSI was found in 9%, ERBB2 amplification in 3%, and POLE pathogenic mutation in 1.5% of patients. Such results mostly parallel frequencies observed in Western patients. However, POLE existed at a higher frequency and was associated with large tumor T-cell infiltration. CONCLUSION: Comparing to the Western counterparts, POLE mutations were increased in our cohort. The prevalence of NTRK gene fusion was around 7% in the MSI-high CRC cohort. Increased adoption of molecular profiling in Asian patients is essential for the improvement of therapeutic outcomes. IMPLICATIONS FOR PRACTICE: The increasing use of genomic profiling assays in colorectal cancer (CRC) has allowed for the identification of a higher number of patient subsets benefiting from matched therapies. With an increase in the number of therapies, assays simultaneously evaluating all candidate biomarkers are critical. The results of this study provide an early support for the feasibility and utility of genomic profiling in Chinese patients with CRC.


Asunto(s)
Neoplasias Colorrectales , ADN Polimerasa II , Inestabilidad de Microsatélites , Proteínas de Unión a Poli-ADP-Ribosa , Receptor ErbB-2 , Receptor trkA , China/epidemiología , Neoplasias Colorrectales/genética , Variaciones en el Número de Copia de ADN , ADN Polimerasa II/genética , Femenino , Genómica , Humanos , Masculino , Persona de Mediana Edad , Mutación , Proteínas de Unión a Poli-ADP-Ribosa/genética , Receptor ErbB-2/genética
5.
Zhonghua Jie He He Hu Xi Za Zhi ; 37(1): 30-5, 2014 Jan.
Artículo en Zh | MEDLINE | ID: mdl-24694971

RESUMEN

OBJECTIVE: To investigate alveolar epithelial cell apoptosis induced by endoplasmic reticulum stress in a rat model of chronic obstructive pulmonary disease (COPD) and the potential protective effect of resveratrol. METHODS: The COPD rat model was established by intratracheal instillation of lipopolysaccharide (LPS) and exposure to cigarette smoke daily. Forty-eight male Sprague-Dawley rats were randomly divided into 4 groups (n = 12 each): a normal control group, a resveratrol control group (resveratrol 25 mg × kg⁻¹ × d⁻¹ gavage), a COPD group (COPD rat model established), and a resveratrol intervention group (COPD model rats receiving resveratrol 25 mg × kg⁻¹ × d⁻¹ gavage). Spirometry was conducted and the lung pathological changes were observed. The protein expression of CCAAT/enhancer binding protein homologous protein (CHOP) and caspase-12 were detected by immunohistochemistry and Western blot, and alveolar epithelial apoptosis was analyzed by terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL). Statistical analysis among groups were carried out by one way analysis of variance followed by LSD-t test between 2 groups. RESULTS: Significant decrease of FEV0.3/FVC [(59 ± 4)%] and dynamic lung compliance [(0.154 ± 0.013) ml/cm H2O, 1 cm H2O = 0.098 kPa] and increase of airway resistance [(0.651 ± 0.046) cm H2O × ml⁻¹× s⁻¹] were found in the COPD group when compared with the normal control group [(82 ± 4)%, (0.401 ± 0.033) ml/cm H2O, (0.404 ± 0.033) cm H2O × ml⁻¹ × s⁻¹] (t = -14.48, 16.48, P < 0.05). The FEV0.3/FVC [(71 ± 5)%] and dynamic lung compliance [(0.302 ± 0.023) ml/cm H2O] of the resveratrol intervention group were significantly improved when compared with those of the COPD group, and the airway resistance [(0.442 ± 0.036) cm H2O × ml⁻¹ × s⁻¹] also decreased (t = -10.02-10.37, P < 0.05). Significant small airway inflammation and emphysema were seen in the lung tissue of COPD group, while significant improvement was observed in the resveratrol intervention group when compared with COPD group. The lung tissue immunohistochemistry integrated optical density (IOD) of CHOP and caspase-12 (9 778 ± 217, 12 009 ± 346) of the COPD group increased significantly when compared with the normal control group (960 ± 94, 1 124 ± 112) (t = -100.43, - 90.43, P < 0.05), while the IODs of the resveratrol intervention group (5 799 ± 177, 6 720 ± 173) decreased significantly when compared with the COPD group (t = 45.32, 43.93, P < 0.05). Western blot results showed that the relative quantification of CHOP (0.910 ± 0.053) and caspase-12 (1.104 ± 0.026) increased in the COPD group when compared with the normal control group (0.204 ± 0.021, 0.133 ± 0.013, t = -36.04, -115.03, P < 0.05), while the ratios of the resveratrol intervention group (0.462 ± 0.037, 0.642 ± 0.011) decreased significantly when compared with COPD group (t = 24.22, 60.59, P < 0.05). Higher apoptosis index was seen in the COPD group [(39.8 ± 1.6)%] when compared with the resveratrol intervention group [(26.3 ± 1.5)%] and the normal control group [(6.4 ± 0.6)%] (t = 20.21, -49.94, P < 0.05). CONCLUSIONS: Endoplasmic reticulum stress, which induced apoptosis of alveolar epithelial cells, was observed in this COPD model. Resveratrol was shown to alleviate endoplasmic reticulum stress and attenuate alveolar epithelial apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico , Pulmón/patología , Enfermedad Pulmonar Obstructiva Crónica/patología , Estilbenos/farmacología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Antioxidantes/farmacología , Caspasa 12/metabolismo , Modelos Animales de Enfermedad , Lipopolisacáridos/efectos adversos , Pulmón/metabolismo , Masculino , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Ratas , Ratas Sprague-Dawley , Resveratrol , Humo/efectos adversos , Factor de Transcripción CHOP/metabolismo
6.
J Cancer Res Clin Oncol ; 149(11): 9007-9016, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37165118

RESUMEN

PURPOSE: Tumor-infiltrating lymphocytes (TILs) have shown remarkable clinical responses in some patients with advanced solid tumors. As a rare subset of TILs, CD4-/CD8- double-negative T cells (DNTs) were poorly known. This study aims to investigate the characteristics and function of CD3+CD4-CD8- TILs (double-negative TIL, DN-TILs) derived from solid tumor. METHODS: DN-TILs were derived and expanded ex vivo from resected gastric carcinoma tissue and phenotyped by flow cytometry. The cytotoxicity of DN-TILs was determined against established tumor cell lines in vitro or through in vivo adoptive transfer into xenograft models. K562 cells were transferred with the HLA gene to verify whether the cytotoxicity of DN-TILs was MHC-independent. RESULTS: Flow cytometric analysis revealed a high-purity population of DN-TILs (> 97%) within CD3+ TILs, which expanded more than 800-folds in 2 weeks, consisting of a mixture of alpha-beta (αß) and gamma-delta (γδ) T-cell receptor (TCR)-expressing cells (with the majority being αß-TCR, > 95%). Using single-cell RNA sequencing, the expanded DN-TILs were categorized into four main subsets, Natural Killer T cells (approximately 80%, 5563 in 7028), Progenitor cells, Germ cells and T helper2 cells. DN-TILs exhibited a broad anticancer cytotoxicity in a donor-unrestricted manner against various cancer cell lines derived from pancreatic cancer (Panc-1), gastric cancer (HGC-27), ovarian cancer (SKOV-3), malignant melanoma (A375). The cytotoxicity was MHC-independent, which was not altered in K562 transferring with HLA gene or not. DN-TILs significantly reduced tumor volume in xenograft models with superior tumor-homing ability and low off-target toxicity. CONCLUSION: Gastric carcinoma derived DN-TIL can target tumor cells in vitro and in vivo. DN-TILs have the potential to be used as a adoptive cell therapy for solid cancers with both the advantages of DNT and TIL.


Asunto(s)
Citotoxicidad Inmunológica , Linfocitos Infiltrantes de Tumor , Humanos , Linfocitos Infiltrantes de Tumor/patología , Línea Celular Tumoral , Linfocitos T CD8-positivos , Receptores de Antígenos de Linfocitos T , Proliferación Celular
7.
Cell Biosci ; 13(1): 65, 2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-36991473

RESUMEN

BACKGROUND: Transmembrane emp24 domain containing (TMED) proteins are known to play pivotal roles in normal development, but have been reported to be implicated in pancreatic disease, immune system disorders, and cancers. As far as TMED3 is concerned, its roles in cancers are controversial. However, evidence describing TMED3 in the context of malignant melanoma (MM) is scarce. RESULTS: In this study, we characterized the functional significance of TMED3 in MM and identified TMED3 as a tumor-promoting factor in MM development. Depletion of TMED3 arrested the development of MM in vitro and in vivo. Mechanistically, we found that TMED3 could interact with Cell division cycle associated 8 (CDCA8). Knocking down CDCA8 suppressed cell events associated with MM development. On the contrary, elevating CDCA8 augmented cell viability and motility and even reversed the inhibitory effects of TMED3 knockdown on MM development. On the other hand, we found that the levels of P-Akt and P-PI3K were decreased in response to TMED3 downregulation, which was partially abolished following SC79 treatment. Thus, our suspicion was that TMED3 exacerbates MM progression via PI3K/Akt pathway. More notably, previously decreased P-Akt and P-PI3K in TMED3-depleted cells were rescued after overexpressing CDCA8. Also, previously impaired cell events due to CDCA8 depletion were ameliorated after SC79 addition, implying that TMED3 regulates PI3K-AKT pathway via CDCA8, thereby promoting MM development. CONCLUSIONS: Collectively, this study established the link between TMED3 and MM, and provides a potential therapeutic intervention for patients with MM harboring abundant TMED3.

8.
Hum Gene Ther ; 34(23-24): 1248-1256, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37917093

RESUMEN

Distant metastasis and primary tumor relapse are the two main hurdles to the success of surgical treatment for cancer patients. Circulating tumor cells (CTCs) and incomplete surgical resection are the primary cause of distant metastasis and local recurrence of tumors, respectively. Chimeric antigen receptor (CAR)-modified T cells target residual carcinomas and CTCs hold the potential to inhibit primary recurrence and reduce tumor metastasis, but the experimental evidence is lacking. Here, we developed a surgery-induced tumor metastasis model in immunocompetent mice to investigate the efficacy of CAR-T cells therapy in preventing metastasis and local recurrence. We observed that subcutaneous tumor resection has induced a large number of CTCs intravasated into circulation. EpCAM-specific CAR-T was effective in clearing CTCs following surgical removal of the tumor. This resulted in less pulmonary metastasis and longer survival in mice when compared to mice treated with surgery followed by Mock-T cells infusion. In addition, the local relapse was obviously inhibited at the surgical site followed by EpCAM-CAR-T cell treatment. This study demonstrated that CAR-T cell therapy can be an adjuvant treatment following surgery to prevent tumor metastasis and inhibit primary tumor relapse for cancer patients.


Asunto(s)
Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , Receptores Quiméricos de Antígenos/genética , Molécula de Adhesión Celular Epitelial/genética , Recurrencia Local de Neoplasia/prevención & control , Recurrencia Local de Neoplasia/patología , Inmunoterapia Adoptiva/métodos , Recurrencia , Tratamiento Basado en Trasplante de Células y Tejidos
9.
Sci Adv ; 9(48): eadg9721, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38039357

RESUMEN

The efficacy of CAR-T cells for solid tumors is unsatisfactory. EpCAM is a biomarker of epithelial tumors, but the clinical feasibility of CAR-T therapy targeting EpCAM is lacking. Here, we report pre- and clinical investigations of EpCAM-CAR-T cells for solid tumors. We demonstrated that EpCAM-CAR-T cells costimulated by Dectin-1 exhibited robust antitumor activity without adverse effects in xenograft mouse models and EpCAM-humanized mice. Notably, in clinical trials for epithelial tumors (NCT02915445), 6 (50%) of the 12 enrolled patients experienced self-remitted grade 1/2 toxicities, 1 patient (8.3%) experienced reversible grade 3 leukopenia, and no higher-grade toxicity reported. Efficacy analysis determined two patients as partial response. Three patients showed >23 months of progression-free survival, among whom one patient experienced 2-year progress-free survival with detectable CAR-T cells 200 days after infusion. These data demonstrate the feasibility and tolerability of EpCAM-CAR-T therapy.


Asunto(s)
Neoplasias Glandulares y Epiteliales , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , Molécula de Adhesión Celular Epitelial , Linfocitos T , Inmunoterapia/efectos adversos , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico
10.
Artículo en Inglés | MEDLINE | ID: mdl-36034960

RESUMEN

Anti-CD19 chimeric antigen receptor (CAR) T-cells are an effective treatment for refractory B-cell lymphoma, but CD19 deletion is prone to relapse. We conducted this study to find more effective dual CAR19/20 T-cells to target B-cell lymphoma and prevent antigen loss leading to recurrence. In this study, we transduced CD19 and CD20 CARs into human T cells in parallel and compared parallel dual CAR19/20, single CAR, and tandem CAR19/20 in vitro and in vivo. After transduction with the corresponding vectors, CD19 and CD20 CARs were dually expressed in human T cells. It was observed that parallel CAR19/20 T-cells contained a substantial proportion of naive subpopulations and were able to proliferate in vitro. Treatment with parallel CAR19/20, single CAR, or tandem CAR19/20 T-cells sustainably induced complete lysis of leukemia cells in a 5 : 1 ratio. Compared with single or tandem CAR T-cell-transplanted mice, parallel CAR19/20 T-cell-transplanted mice exhibited smaller tumor volume, more stable body weight, and longer survival. This suggests that parallel CAR19/20 has superior antilymphoma activity in vivo. In addition, parallel CAR19/20 T-cells were also able to kill patients' lymphoma cells in vitro. Therefore, it can be considered that parallel CAR19/20 is equally effective against single CAR and tandem CAR19/20 in vitro but more effective against lymphoma cells in vivo. This is a promising treatment to prevent the recurrence of antigen loss following CD19-targeted therapy in B lymphoma.

11.
J Cell Biochem ; 112(11): 3406-20, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21769915

RESUMEN

Oxygen deficiency and nutrient deprivation widely exists in solid tumors because of the poor blood supply. However, cancer cells can survive this adverse condition and proliferate continuously to develop. To figure out the way to survive, we investigated the role of autophagy in the microenvironment in hepatocellular carcinoma. In order to simulate the tumor microenvironment more veritably, cells were cultured in oxygen-nutrient-deprived condition following a hypoxia preconditioning. As a result, cell death under hypoxia plus nutrient deprivation was much less than that under nutrient deprivation only. And the decreased cell death mainly attributed to the decreased apoptosis. GFP-LC3 and electron microscopy analysis showed that autophagy was significantly activated in the period of hypoxia preconditioning. However, autophagic inhibitor-3-MA significantly abrogated the apoptosis reduction in hypoxia, which implied the involvement of autophagy in protection of hepatocellular carcinoma cells against apoptosis induced by starvation. Furthermore, Beclin 1 was proved to play an important role in this process. siRNA targeting Beclin 1 was transfected into hepatocellular carcinoma cells. And both data from western blot detecting the expression of LC3-II and transmission microscopy observing the accumulation of autophagosomes showed that autophagy was inhibited obviously as a result of Beclin 1 knockdown. Besides, the decreased apoptosis of starved cells under hypoxia was reversed. Taken together, these results suggest that autophagy activated by hypoxia mediates the tolerance of hepatocellular carcinoma cells to nutrient deprivation, and this tolerance is dependent on the activity of Beclin 1.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis , Autofagia , Carcinoma Hepatocelular/patología , Hipoxia de la Célula , Neoplasias Hepáticas/patología , Proteínas de la Membrana/fisiología , Beclina-1 , Western Blotting , Línea Celular Tumoral , Humanos , Microscopía Electrónica de Transmisión , ARN Interferente Pequeño
13.
Front Oncol ; 11: 683502, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34692475

RESUMEN

INTRODUCTION: Anlotinib (AL3818) is a novel multi-target tyrosine kinase inhibitor (TKI) targeting vascular endothelial growth factor receptor (VEGFR) and suppressing tumor growth. Modulation of tumor suppressive immune microenvironment via the inhibition of vascular endothelial growth factor may augment the activity of immune checkpoint inhibitors. Here we described the results of safety, and clinical efficacy of anlotinib combined with immunotherapy in patients with advanced solid tumors, the serum cytokine levels, and peripheral blood T lymphocyte populations were detected simultaneously. METHODS: Twenty six cases with advanced late-stage cancers including lung, gallbladder, endometrial, gastric, pancreatic, penile cancers and melanoma were treated since January 2019. Patients received a combination of anlotinib (12mg) once daily on day 1 to day 14 (21 days as a course) plus anti-PD-1 antibodies every 3 weeks until progression or intolerable toxicity. Imaging was performed every 6 weeks for the first year of therapy. Blood samples were collected from patients prospectively. Serum interleukin (IL)-2, IL-4, IL-6, IL-10, Tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ) and circulating immune cell subsets were measured at baseline and after two cycles of treatment via flow cytometry. RESULTS: There were ten tumor types enrolled with lung, gallbladder, cholangiocarcinoma and soft tissue sarcoma being the most common. Most patients had received front line treatments for metastatic disease (80.8%). The objective response rate (ORR) was 23.1%, including one complete response (CR) (3.8%) and five partial responses (PR) (19.2%) and a disease control rate (DCR=CR+PR+SD) of 80.8% (21 of 26). The median PFS was 8.37 months (95% CI: 6.5-10.0 months). Three patients (11.5%) had grade 3 treatment-related adverse events. There were no grade 4 or 5 treatment-related adverse events. Grades 3 toxicities included hand-foot syndrome (n=2) and hypertension (n=1). Higher serum IL-2, IL-4, IL-10, TNF-α, IFN-γ levels and lower ratios of CD4/CD8 T cells were found in the responders compared with non-responders. CONCLUSIONS: The preliminary data showed that the combination of anlotinib and anti-PD-1 antibodies demonstrated promising durable antitumor efficacy with acceptable toxicity in patients with various advance tumors, and promoted favorable changes in serum IL-2, IL-4, IL-10, TNF-α, IFN-γ levels and circulating immune cell subsets in clinical responders. It is worth to further validate the efficacy in a randomized prospective trial.

14.
Signal Transduct Target Ther ; 6(1): 398, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34795206

RESUMEN

Given that only a subset of patients with colorectal cancer (CRC) benefit from immune checkpoint therapy, efforts are ongoing to identify markers that predict immunotherapeutic response. Increasing evidence suggests that microbes influence the efficacy of cancer therapies. Fusobacterium nucleatum induces different immune responses in CRC with different microsatellite-instability (MSI) statuses. Here, we investigated the effect of F. nucleatum on anti-PD-L1 therapy in CRC. We found that high F. nucleatum levels correlate with improved therapeutic responses to PD-1 blockade in patients with CRC. Additionally, F. nucleatum enhanced the antitumor effects of PD-L1 blockade on CRC in mice and prolonged survival. Combining F. nucleatum supplementation with immunotherapy rescued the therapeutic effects of PD-L1 blockade. Furthermore, F. nucleatum induced PD-L1 expression by activating STING signaling and increased the accumulation of interferon-gamma (IFN-γ)+ CD8+ tumor-infiltrating lymphocytes (TILs) during treatment with PD-L1 blockade, thereby augmenting tumor sensitivity to PD-L1 blockade. Finally, patient-derived organoid models demonstrated that increased F. nucleatum levels correlated with an improved therapeutic response to PD-L1 blockade. These findings suggest that F. nucleatum may modulate immune checkpoint therapy for CRC.


Asunto(s)
Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Colorrectales , Fusobacterium nucleatum/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteínas de Neoplasias/inmunología , Animales , Células CACO-2 , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Humanos , Ratones
15.
Mol Ther Nucleic Acids ; 24: 923-938, 2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34094711

RESUMEN

Cancer cachexia is a kind of whole-body metabolic disorder syndrome accompanied by severe wasting of muscle tissue in which cancer exosomes may be involved. Analysis of clinical samples showed that the serum exosome concentrations were correlated with the development of cancer cachexia. Exosomes secreted by C26 cells could decrease the diameter of C2C12 myotubes in vitro and decrease mouse muscle strength and tibialis anterior (TA) muscle weight in vivo. GW4869, an inhibitor of exosome excretion, ameliorated muscle wasting in C26 tumor-bearing mice. MicroRNA (miRNA) sequencing (miRNA-seq) analysis suggested that miR-195a-5p and miR-125b-1-3p were richer in C26 exosomes than in exosomes secreted from MC38 cells (non-cachexic). Both miR-195a-5p and miR-125b-1-3p mimics could induce atrophy of C2C12 myoblasts. Downregulation of Bcl-2 and activation of the apoptotic signaling pathway were observed in C2C12 myoblasts transfected with miR-195a-5p and miR-125b-1-3p mimics, in the gastrocnemius muscle of C26 tumor-bearing mice and in the TA muscle injected with C26 exosomes. Results of dual-luciferase assay confirmed the targeting of miR-195a-5p/miR-125b-1-3p to Bcl-2. Overexpression of Bcl-2 successfully reversed atrophy of C2C12 myoblasts induced by the two miRNA mimics. These results suggested that cancer exosome enriched miRNAs might induce muscle atrophy by targeting Bcl-2-mediated apoptosis.

16.
Transl Oncol ; 14(10): 101168, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34252743

RESUMEN

Genomic aberrations (GAs) in fibroblast growth factor receptors (FGFRs) are involved in the pathogenesis of intrahepatic cholangiocarcinoma (ICC), and clinical trials have shown efficacy of FGFR inhibitors in treating ICC patients with FGFR GAs such as FGFR2 rearrangement. To clarify the FGFRs GA profile and corresponding clinicopathological features in Chinese patients with ICC, a total of 257 cases were identified. Fourteen cases (5.45%) were positive for FGFR2 rearrangement. Further analysis on the 110 FGFR2 rearrangement negative cases showed that 13 patients present additional FGFRs GAs, including FGFR3 rearrangement (2.73%), and FGFRs mutations. When compared with patients without FGFRs GAs, those with FGFR2 or FGFR3 rearrangement presented more under the age of 58 years, female sex, HBsAb positivity, CD10 expression, and PD-L1 expression. The clinical characteristics between patients with FGFRs mutation and those without FGFRs GAs were similar, with the exception that cases with FGFRs mutation have more hepatolithiasis. We concluded that FGFR rearrangement is associated with unique clinical phenotypes in ICC.

17.
Signal Transduct Target Ther ; 6(1): 26, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33473101

RESUMEN

Neoantigens are considered to be ultimate target of tumor immunotherapy due to their high tumor specificity and immunogenicity. Dendritic cell (DCs) vaccines based on neoantigens have exciting effects in treatment of some malignant tumors and are a promising therapeutic modality. Lung cancer is a lethal disease with the highest morbidity and mortality rate in the world. Despite the rapid development of targeted therapy and immune checkpoint inhibitors for lung cancer in recent years, their efficacy is still unsatisfactory overall. Therefore, there is an urgent unmet clinical need for lung cancer treatment. Here, we attempted to treat lung cancer using a personalized neoantigen peptide-pulsed autologous DC vaccine and conducted a single-arm, 2 medical centers, pilot study initiated by the investigator (ChiCTR-ONC-16009100, NCT02956551). The patients enrolled were patients with heavily treated metastatic lung cancer. Candidate neoantigens were derived from whole-exome sequencing and RNA sequencing of fresh biopsy tissues as well as bioinformatics analysis. A total of 12 patients were enrolled in this study. A total of 85 vaccine treatments were administered with a median value of 5 doses/person (range: 3-14 doses/person). In total, 12-30 peptide-based neoantigens were selected for each patient. All treatment-related adverse events were grade 1-2 and there were no delays in dosing due to toxic effects. The objective effectiveness rate was 25%; the disease control rate was 75%; the median progression-free survival was 5.5 months and the median overall survival was 7.9 months. This study provides new evidence for neoantigen vaccine therapy and new therapeutic opportunities for lung cancer treatment.


Asunto(s)
Antígenos de Neoplasias , Vacunas contra el Cáncer , Células Dendríticas , Inmunoterapia , Neoplasias Pulmonares , Medicina de Precisión , Anciano , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Autoinjertos , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Supervivencia sin Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Proyectos Piloto , Tasa de Supervivencia
18.
Exp Mol Pathol ; 88(1): 112-7, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19879258

RESUMEN

CpG island methylator phenotype (CIMP), in which multiple genes are concurrently methylated, is an important mechanism in hepatocellular carcinoma development. We determined a hypermethylation profile in hepatocellular carcinoma (HCC). We examined the promoter methylation status of 10 genes in 60 cases of hepatocellular carcinoma (HCC), 60 cases of paired non-tumor tissues, and 6 cases of normal tissues by methylation-specific PCR. The average methylated gene numbers were significantly different between HCC and nontumor tissues (p<0.001). We found metastasis, gamma-glutamyl transpeptidase (GGT) and tumor node metastasis (TNM) stage were significantly different among patients with different CIMP status. Patients with high frequency CIMP tumors had significantly worse survival than patients with intermediate frequency or no CIMP tumors (p<0.01 and p<0.05, respectively). Our results suggested that CIMP could serve as a molecular marker of late stage and poorly prognostic HCC development.


Asunto(s)
Carcinoma Hepatocelular/genética , Islas de CpG/genética , Neoplasias Hepáticas/genética , Regiones Promotoras Genéticas/genética , Adulto , Anciano , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/secundario , Metilación de ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Fenotipo , Pronóstico , Tasa de Supervivencia , gamma-Glutamiltransferasa/metabolismo
19.
Cell Death Dis ; 11(7): 496, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32612115

RESUMEN

Chondroitin polymerizing factor (CHPF) is an important member of glycosyltransferases involved in the biosynthesis of chondroitin sulfate (CS). However, the relationship between CHPF and malignant melanoma (MM) is still unknown. In this study, it was demonstrated that CHPF was up-regulated in MM tissues compared with the adjacent normal skin tissues and its high expression was correlated with more advanced T stage. Further investigations indicated that the over-expression/knockdown of CHPF could promote/inhibit proliferation, colony formation and migration of MM cells, while inhibiting/promoting cell apoptosis. Moreover, knockdown of CHPF could also suppress tumorigenicity of MM cells in vivo. RNA-sequencing followed by Ingenuity pathway analysis (IPA) was performed for exploring downstream of CHPF and identified CDK1 as the potential target. Furthermore, our study revealed that knockdown of CDK1 could inhibit development of MM in vitro, and alleviate the CHPF over-expression induced promotion of MM. In conclusion, our study showed, as the first time, CHPF as a tumor promotor for MM, whose function was carried out probably through the regulation of CDK1.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Melanoma/metabolismo , Melanoma/patología , Apoptosis , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Melanoma/enzimología , Melanoma/genética , Persona de Mediana Edad , N-Acetilgalactosaminiltransferasas , Piel/metabolismo , Piel/patología
20.
Oncol Rep ; 22(3): 549-56, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19639202

RESUMEN

The telomerase is specifically activated in most malignant tumors but is usually inactive in normal somatic cells. It has been reported that telomerase has an anti-apoptotic role and up-regulation of telomerase helps cancer cells to be resistant to chemotherapeutic agent-induced cell death. The effect of cisplatin on telomerase activity is complex, and the exact mechanism remains largely unknown. In this study, we found that cisplatin activated telomerase activity and human telomerase reverse transcriptase (hTERT) expression in SMMC7721 human hepatocellular carcinoma cell line. Low-dose cisplatin up-regulated hTERT and NF-kappaB p65 expression and increased telomerase and NF-kappaB activity. Inhibition of NF-kappaB attenuated the hTERT expression and telomerase activity exposed to cisplatin, suggesting that NF-kappaB is responsible for the cisplatin-induced activation of the hTERT. Furthermore, preincubation of low-dose cisplatin which induced high expression of hTERT help hepatocellular carcinoma SMMC7721 cells survive under the high concentration of anti-cancer drugs. Inhibition of hTERT increased sensitivity of SMMC7721 cells to chemotherapy. Taken together, these results suggested that up-regulation of hTERT expression by low-dose cisplatin is NF-kappaB-dependent and contributes to chemotherapy resistance in human hepatocellular cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Telomerasa/genética , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Daño del ADN , Resistencia a Antineoplásicos , Regulación Enzimológica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , FN-kappa B/fisiología , Telomerasa/análisis , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA