Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Adv Mater ; 36(15): e2304328, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38229577

RESUMEN

Currently, certain cancer patients exhibit resistance to radiotherapy due to reduced DNA damage under hypoxic conditions and acquired immune tolerance triggered by transforming growth factor-ß1 (TGF-ß1) and membrane-localized programmed death ligand-1 (PD-L1). Meanwhile, cytoplasm-distributed PD-L1 induces radiotherapy resistance through accelerating DNA damage repair (DDR). However, the disability of clinically used PD-L1 antibodies in inhibiting cytoplasm-distributed PD-L1 limits their effectiveness. Therefore, a nanoadjuvant is developed to sensitize cancer to radiotherapy via multi-level immunity activation through depressing PD-L1 and TGF-ß1 by triphenylphosphine-derived metformin, and activating the cGAS-STING pathway by generating Mn2+ from MnO2 and producing more dsDNA via reversing tumor hypoxia and impairing DDR. Thus, Tpp-Met@MnO2@Alb effectively enhances the efficiency of radiotherapy to inhibit the progression of irradiated local and abscopal tumors and tumor lung metastases, offering a long-term memory of antitumor immunity without discernible side effects. Overall, Tpp-Met@MnO2@Alb has the potential to be clinically applied for overcoming radio-immunotherapy resistance.


Asunto(s)
Adyuvantes Farmacéuticos , Neoplasias Pulmonares , Neoplasias , Humanos , Antígeno B7-H1/antagonistas & inhibidores , Inmunoterapia , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/terapia , Compuestos de Manganeso/farmacología , Neoplasias/radioterapia , Neoplasias/terapia , Óxidos , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Adyuvantes Farmacéuticos/farmacología , Adyuvantes Farmacéuticos/uso terapéutico , Nucleotidiltransferasas/efectos de los fármacos , Proteínas de la Membrana/efectos de los fármacos
2.
Integr Cancer Ther ; 23: 15347354231226108, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38240227

RESUMEN

OBJECTIVE: In China, grade 2 to 3 immune-related rash will probably lead to the interruption of immunotherapy. Corticosteroid (CS) is the main treatment, but not always effective. The external application of clearing heat and removing dampness, which is represented by Qing-Re-Li-Shi Formula (QRLSF), has been used in our hospital to treat immune-related cutaneous adverse events (ircAEs) for the last 5 years. The purpose of this study was to discuss its efficacy and safety in the treatment of grade 2 to 3 rash. METHODS: A retrospective study of patients with grade 2 to 3 immune-related rash in our hospital from December 2019 to December 2022 was conducted. These patients received QRLSF treatment. Clinical characteristics, treatment outcome, and health-related quality of life (HrQoL) were analyzed. RESULTS: Thirty patients with grade 2 to 3 rash (median onset time: 64.5 days) were included. The skin lesions of 24 cases (80%) returned to grade 1 with a median time of 8 days. The accompanying symptoms were also improved with median time of 3 to 4 days. The addition of antihistamine (AH) drug didn't increase the efficacy of QRLSF (AH + QRLSF: 75.00% vs QRLSF: 83.33%, P = .66). No significant difference was observed in the efficacy of QRLSF treatment regardless of whether patients had previously received CS therapy (untreated population: 88.24% vs treated population: 69.23%, P = .36). During 1-month follow-up, 2 cases (8.33%) underwent relapses. In terms of HrQoL, QRLSF treatment could significantly reduce the median scores of all domains of Skindex-16, including symptoms (39.58 vs 8.33, P < .0001), emotions (58.33 vs 15.48, P < .0001), functioning (46.67 vs 13.33, P < .0001) and composite (52.60 vs 14.06, P < .0001). CONCLUSION: External application of clearing heat and removing dampness was proven to be an effective and safe treatment for such patients. In the future, high-quality trials are required to determine its clinical application in the field of ircAEs.


Asunto(s)
Antígeno B7-H1 , Exantema , Receptor de Muerte Celular Programada 1 , Humanos , Antígeno B7-H1/antagonistas & inhibidores , Exantema/inducido químicamente , Exantema/tratamiento farmacológico , Calor , Ligandos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Calidad de Vida , Estudios Retrospectivos
3.
Cell Death Dis ; 14(2): 170, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36854755

RESUMEN

Patients with advanced melanoma have shown an improved outlook after anti-PD1 therapy, but the low response rate restricts clinical benefit; therefore, enhancing anti-PD1 therapeutic efficacy remains a major challenge. Here, our findings showed a significantly increased abundance of α-KG in healthy controls, anti-PD1-sensitive melanoma-bearing mice, and anti-PD1-sensitive melanoma patients; moreover, supplementation with α-KG enhanced the efficacy of anti-PD1 immunotherapy and increased PD-L1 expression in melanoma tumors via STAT1/3. We also found that supplementation with α-KG significantly increased the activity of the methylcytosine dioxygenases TET2/3, which led to an increased 5-hydroxymethylcytosine (5-hmC) level in the PD-L1 promoter. As a consequence, STAT1/3 binding to the PD-L1 promoter was stabilized to upregulate PD-L1 expression. Importantly, single-cell sequencing of preclinical samples and analysis of clinical data revealed that TET2/3-STAT1/3-CD274 signaling was associated with sensitivity to anti-PD1 treatment in melanoma. Taken together, our results provide novel insight into α-KG's function in anti-PD1 treatment of melanoma and suggest supplementation with α-KG as a novel promising strategy to improve the efficacy of anti-PD1 therapy.


Asunto(s)
Antígeno B7-H1 , Ácidos Cetoglutáricos , Melanoma , Animales , Ratones , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Suplementos Dietéticos , Epigénesis Genética , Ácidos Cetoglutáricos/farmacología , Ácidos Cetoglutáricos/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/genética
4.
Phytother Res ; 37(5): 1740-1753, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36576358

RESUMEN

The efficacy of anti-PD-1 therapy is not as expected in hepatocellular carcinoma (HCC). YAP1 was overexpressed and activated in HCC. The mechanism of YAP1 in HCC immune escape is unclear. Anti-PD-1 treatment increased YAP1 expression in liver tumor cells, and exhausted CD4+ and CD8+ T cells in the blood and spleen of liver tumor mice. YAP1 knockdown suppressed PD-L1 expression, which was involved in JAK1/STAT1, 3 pathways. Moreover, Yap1 knockout elevated CD4+ and CD8+ T cells in liver tumor niche. Consistently, verteporfin, YAP1 inhibitor, decreased TGF-ß and IFN-γ in liver tumor niche and exhausted CD8+ T cell in the spleen. DHA suppressed YAP1 expression and break immune evasion in liver tumor niche, characterized by decreased PD-L1 in liver tumor cells and increased CD8+ T cell infiltration. Furthermore, DHA combined with anti-PD-1 treatment promoted CD4+ T cell infiltration in the spleen and CD8+ T cell in tumor tissues of mice. In summary, YAP1 knockdown in liver tumor cells suppressed PD-L1 expression and recruited cytotoxic T lymphocytes (CTLs), leading to break immune evasion in tumor niche. Mechanistically, YAP1 knockdown suppressed PD-L1 expression, which was involved in JAK1/STAT1, 3 pathways. Finally, DHA inhibited YAP1 expression, which not only inhibited liver tumor proliferation but also break the immunosuppressive niche in liver tumor tissues and improve the effect of anti-PD-1 therapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Ratones , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Linfocitos T CD8-positivos , Inmunosupresores , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Microambiente Tumoral , Proteínas Señalizadoras YAP/efectos de los fármacos , Proteínas Señalizadoras YAP/genética
5.
Biochem Pharmacol ; 197: 114940, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35120895

RESUMEN

Programmed death ligand-1 (PD-L1) and indoleamine 2, 3-dioxygenase 1 (IDO1) are immune checkpoints induced by interferon-γ (IFN-γ) in the tumor microenvironment, leading to immune escape of tumors. Myricetin (MY) is a flavonoid distributed in many edible and medicinal plants. In this study, MY was identified to inhibit IFN-γ-induced PD-L1 expression in human lung cancer cells. It also reduced the expression of IDO1 and the production of kynurenine which is the product catalyzed by IDO1, while didn't show obvious effect on the expression of major histocompatibility complex-I (MHC-I), a crucial molecule for antigen presentation. In addition, the function of T cells was evaluated using a co-culture system consist of lung cancer cells and the Jurkat-PD-1 T cell line overexpressing PD-1. MY restored the survival, proliferation, CD69 expression and interleukin-2 (IL-2) secretion of Jurkat-PD-1 T cells suppressed by IFN-γ-treated lung cancer cells. Mechanistically, IFN-γ up-regulated PD-L1 and IDO1 at the transcriptional level through the JAK-STAT-IRF1 axis, which was targeted and inhibited by MY. Together, our research revealed a new mechanism of MY mediated anti-tumor activity and highlighted the potential implications of MY in tumor immunotherapy.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Interferón gamma/farmacología , Neoplasias Pulmonares/metabolismo , Células A549 , Antígeno B7-H1/biosíntesis , Antígeno B7-H1/genética , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/fisiología , Células HCT116 , Células HEK293 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Células Jurkat , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología
6.
Phytomedicine ; 96: 153913, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35026515

RESUMEN

BACKGROUND: Anti-PD-1 was used to treat for many cancers, but the overall response rate of monoclonal antibodies blocking the inhibitory PD-1/PD-L1 was less than 20%. Lipid droplet (LD) deposition reduced chemotherapy efficacy, but whether LD deposition affects anti-PD-1 treatment and its mechanism remains unclear. Dihydroartemisinin (DHA) was FDA proved antimalarial medicine, but its working mechanism on LD deposition has not been clarified. PURPOSE: This study aimed to elucidate the mechanism of DHA reducing LDs deposition and improving the efficacy of anti-PD-1. METHODS: LD numbers and area were separately detected by electron microscopy and oil Red O staining. The expression of YAP1 and PLIN2 was detected by immunohistochemical staining in liver cancer tissues. Transcription and protein expression levels of YAP1 and PLIN2 in cells were detected by qRT-PCR and Western blot after DHA treated HepG2215 cells and Yap1LKO mice. RESULTS: LD accumulation was found in the liver tumor cells of DEN/TOPBCOP-induced liver tumor mice with anti-PD-1 treatment. But DHA treatment or YAP1 knockdown reduced LD deposition and PLIN2 expression in HepG2215 cells. Furthermore, DHA reduced the LD deposition, PLIN2 expression and triglycerides (TG) content in the liver tumor cells of Yap1LKO mice with liver tumor. CONCLUSION: Anti-PD-1 promoted LD deposition, while YAP1 knockdown/out reduced LD deposition in HCC. DHA reduced LD deposition by inhibiting YAP1, enhancing the effect of anti-PD-1 therapy.


Asunto(s)
Antígeno B7-H1 , Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Artemisininas , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma Hepatocelular/tratamiento farmacológico , Células Hep G2 , Humanos , Gotas Lipídicas , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Ratones Noqueados , Perilipina-2 , Proteínas Señalizadoras YAP
7.
Eur J Pharmacol ; 916: 174726, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34954232

RESUMEN

Chemotherapy is commonly used in the clinical treatment of melanoma, but it is prone to resistance leading to the poor effectiveness. The mechanisms of resistance are complicated including the cancer stemness. Astragalus polysaccharide (APS) is one of the active components of traditional Chinese herbal medicine Astragalus Membranaceus. Our previous work was reported that APS had an inhibitory effect on the stemness of melanoma. In this study we established chemo-resistant melanoma cells and found that expression of stemness genes were upregulated in the resistant melanoma cells. And APS could downregulate expression of stemness genes. Furthermore, APS combined with cisplatin (DDP) could significantly slow down the tumor growth in the mouse model induced by DDP-resistant cells. In addition, we found that programmed death-ligand 1 (PD-L1) expression could be downregulated and the PI3K/AKT signaling could be affected by APS. These results suggested that APS could be a potential candidate in combination with chemotherapeutic agents, which might play a role in reducing the occurrence of resistance and improving the prognosis of melanoma patients.


Asunto(s)
Planta del Astrágalo , Antígeno B7-H1 , Resistencia a Antineoplásicos , Medicamentos Herbarios Chinos , Melanoma , Polisacáridos , Animales , Planta del Astrágalo/química , Antígeno B7-H1/antagonistas & inhibidores , Regulación hacia Abajo , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Melanoma/tratamiento farmacológico , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Polisacáridos/farmacología , Polisacáridos/uso terapéutico
8.
J Nanobiotechnology ; 19(1): 433, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930269

RESUMEN

BACKGROUND: The construction of a nanoimmune controlled-release system that spatiotemporally recognizes tumor lesions and stimulates the immune system response step by step is one of the most potent cancer treatment strategies for improving the sensitivity of immunotherapy response. RESULTS: Here, a composite nanostimulator (CNS) was constructed for the release of second near-infrared (NIR-II) photothermal-mediated immune agents, thereby achieving spatiotemporally controllable photothermal-synergized immunotherapy. CNS nanoparticles comprise thermosensitive liposomes as an outer shell and are internally loaded with a NIR-II photothermal agent, copper sulfide (CuS), toll-like receptor-9 (TLR-9) agonist, cytosine-phospho-guanine oligodeoxynucleotides, and programmed death-ligand 1 (PD-L1) inhibitors (JQ1). Following NIR-II photoirradiation, CuS enabled the rapid elevation of localized temperature, achieving tumor ablation and induction of immunogenic cell death (ICD) as well as disruption of the lipid shell, enabling the precise release of two immune-therapeutical drugs in the tumor region. Combining ICD, TLR-9 stimulation, and inhibited expression of PD-L1 allows the subsequent enhancement of dendritic cell maturation and increases infiltration of cytotoxic T lymphocytes, facilitating regional antitumor immune responses. CONCLUSION: CNS nanoparticle-mediated photothermal-synergized immunotherapy efficiently suppressed the growth of primary and distant tumors in two mouse models and prevented pulmonary metastasis. This study thus provides a novel sight into photo-controllably safe and efficient immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Rayos Infrarrojos , Nanopartículas/química , Neoplasias/terapia , Fototerapia/métodos , Animales , Azepinas/química , Azepinas/farmacología , Azepinas/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Cobre/química , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Verde de Indocianina/química , Verde de Indocianina/uso terapéutico , Liposomas/química , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Receptor Toll-Like 9/metabolismo , Trasplante Heterólogo , Triazoles/química , Triazoles/farmacología , Triazoles/uso terapéutico
9.
Front Immunol ; 12: 737076, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659228

RESUMEN

Immune checkpoints such as programmed death-1 (PD-1) have been proven as antitumor targets by enhancing cytotoxic T cell activity. All immune checkpoint blockades are antibody therapeutics that have large size and high affinity, as well as known immune-related side effects and low responses. To overcome the limitation of antibody therapeutics, we have explored PD-1/PD-L1 (programmed death-ligand 1) blockades in traditional oriental medicine, which has a long history but has not yet studied PD-1/PD-L1 blockades. Sanguisorbae Radix extract (SRE) blocked PD-1 and PD-L1 binding in competitive ELISA. SRE effectively inhibited the PD-1/PD-L1 interaction, thereby improving T cell receptor (TCR) signaling and the NFAT-mediated luciferase activity of T cells. SRE treatment reduced tumor growth in the humanized PD-L1 MC38 cell allograft humanized PD-1 mouse model. Additionally, the combination of SRE and pembrolizumab (anti-PD-1 antibody) suppressed tumor growth and increased infiltrated cytotoxic T cells to a greater extent did either agent alone. This study showed that SRE alone has anticancer effects via PD-1/PD-L1 blockade and that the combination therapy of SRE and pembrolizumab has enhanced immuno-oncologic effects.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Extractos Vegetales/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Sanguisorba , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células CHO , Técnicas de Cocultivo , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Cricetulus , Humanos , Células Jurkat , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Extractos Vegetales/aislamiento & purificación , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Sanguisorba/química , Transducción de Señal , Carga Tumoral
10.
Front Endocrinol (Lausanne) ; 12: 712107, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34475850

RESUMEN

Background: Treatment options for poorly differentiated (PDTC) and anaplastic (ATC) thyroid carcinoma are unsatisfactory and prognosis is generally poor. Lenvatinib (LEN), a multi-tyrosine kinase inhibitor targeting fibroblast growth factor receptors (FGFR) 1-4 is approved for advanced radioiodine refractory thyroid carcinoma, but response to single agent is poor in ATC. Recent reports of combining LEN with PD-1 inhibitor pembrolizumab (PEM) are promising. Materials and Methods: Primary ATC (n=93) and PDTC (n=47) tissue samples diagnosed 1997-2019 at five German tertiary care centers were assessed for PD-L1 expression by immunohistochemistry using Tumor Proportion Score (TPS). FGFR 1-4 mRNA was quantified in 31 ATC and 14 PDTC with RNAscope in-situ hybridization. Normal thyroid tissue (NT) and papillary thyroid carcinoma (PTC) served as controls. Disease specific survival (DSS) was the primary outcome variable. Results: PD-L1 TPS≥50% was observed in 42% of ATC and 26% of PDTC specimens. Mean PD-L1 expression was significantly higher in ATC (TPS 30%) than in PDTC (5%; p<0.01) and NT (0%, p<0.001). 53% of PDTC samples had PD-L1 expression ≤5%. FGFR mRNA expression was generally low in all samples but combined FGFR1-4 expression was significantly higher in PDTC and ATC compared to NT (each p<0.001). No impact of PD-L1 and FGFR 1-4 expression was observed on DSS. Conclusion: High tumoral expression of PD-L1 in a large proportion of ATCs and a subgroup of PDTCs provides a rationale for immune checkpoint inhibition. FGFR expression is low thyroid tumor cells. The clinically observed synergism of PEM with LEN may be caused by immune modulation.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos , Antineoplásicos Inmunológicos , Antígeno B7-H1/análisis , Evaluación Preclínica de Medicamentos/métodos , Femenino , Alemania , Humanos , Masculino , Persona de Mediana Edad , Compuestos de Fenilurea/farmacología , Quinolinas/farmacología , ARN Mensajero/análisis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Carcinoma Anaplásico de Tiroides/química , Carcinoma Anaplásico de Tiroides/patología , Neoplasias de la Tiroides/química , Neoplasias de la Tiroides/patología
11.
Cancer Lett ; 522: 238-254, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34571084

RESUMEN

The response rate of anti-PD therapy in most cancer patients remains low. Therapeutic drug and tumor-infiltrating lymphocytes (TILs) are usually obstructed by the stromal region within tumor microenvironment (TME) rather than distributed around tumor cells, thus unable to induce the immune response of cytotoxic T cells. Here, we constructed the cationic thermosensitive lipid nanoparticles IR780/DPPC/BMS by introducing cationic NIR photosensitizer IR-780 iodide (IR780) modified lipid components, thermosensitive lipid DPPC and PD-1/PD-L1 inhibitor BMS202 (BMS). Upon laser irradiation, IR780/DPPC/BMS penetrated into deep tumor, and reduced cancer-associated fibroblasts (CAFs) around tumor cells to remodel the spatial distribution of TILs in TME. Interestingly, the cationic IR780/DPPC/BMS could capture released tumor-associated antigens (TAAs), thereby enhancing the antigen-presenting ability of DCs to activate cytotoxic T lymphocytes. Moreover, IR780/DPPC/BMS initiated gel-liquid crystal phase transition under laser irradiation, accelerating the disintegration of lipid bilayer structure and leading to the responsive release of BMS, which would reverse the tumor immunosuppression state by blocking PD-1/PD-L1 pathway for a long term. This combination treatment can synergistically exert the antitumor immune response and inhibit the tumor growth and metastasis.


Asunto(s)
Antígeno B7-H1/inmunología , Liposomas/farmacología , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/inmunología , Acetamidas/química , Acetamidas/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunoterapia/métodos , Indoles/química , Indoles/farmacología , Liposomas/química , Terapia por Luz de Baja Intensidad , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Nanopartículas/química , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/radioterapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Piridinas/química , Piridinas/farmacología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/efectos de la radiación , Microambiente Tumoral/efectos de los fármacos
12.
Biochem Biophys Res Commun ; 561: 187-194, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34023785

RESUMEN

Interaction of programmed death-ligand 1 (PD-L1) and programmed death-1 (PD-1) inhibits T cell activation. Tumor tissues can evade immune surveillance by expressing higher levels of PD-L1. Identification of potential regulators of PD-L1 through natural metabolites may contribute to discovering new drugs for immunotherapy. By using a metabolite library screen, we showed that pyridoxal (PL) significantly suppresses PD-L1 expression. Mechanistically, PL accelerates PD-L1 degradation in a proteasome-dependent manner, and STUB1 serves as an E3 ligase during the process. Functionally, PL enhances T cell killing activity by blocking the PD-1/PD-L1 signaling pathway. Thus, we have identified PL as an inhibitor of PD-L1, which provides a feasible option for combination immunotherapy.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Inmunoterapia/métodos , Neoplasias/inmunología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Vitamina B 6/farmacología , Antígeno B7-H1/inmunología , Células Cultivadas , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteolisis , Transducción de Señal , Complejo Vitamínico B/farmacología
13.
Phytother Res ; 35(7): 3916-3935, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33970512

RESUMEN

The programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway is abnormally expressed in cervical cancer cells. Moreover, PD-1/PD-L1 blockade reduces the apoptosis and exhaustion of T cells and inhibits the development of malignant tumors. Usnic acid is a dibenzofuran compound originating from Usnea diffracta Vain and has anti-inflammatory, antifungal, and anticancer activities. However, the molecular mechanism of its antitumor effects has not been fully elucidated. In this work, we first observed that usnic acid decreased the expression of PD-L1 in HeLa cells and enhanced the cytotoxicity of co-cultured T cells toward tumor cells. Usnic acid inhibited PD-L1 protein synthesis by reducing STAT3 and RAS pathways cooperatively. It was subsequently shown that usnic acid induced MiT/TFE nuclear translocation through the suppression of mTOR signaling pathways, and promoted the biogenesis of lysosomes and the translocation of PD-L1 to the lysosomes for proteolysis. Furthermore, usnic acid inhibited cell proliferation, angiogenesis, migration, and invasion, respectively, by downregulating PD-L1, thereby inhibiting tumor growth. Taken together, our results show that usnic acid is an effective inhibitor of PD-L1 and our study provide novel insights into the mechanism of its anticancer targeted therapy.


Asunto(s)
Antígeno B7-H1 , Benzofuranos/farmacología , Proliferación Celular/efectos de los fármacos , Linfocitos T/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Células HeLa , Humanos , Parmeliaceae/química
14.
J Med Chem ; 64(9): 5519-5534, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33938739

RESUMEN

Through specific structural modification of a 4-phenylindoline precursor, new 4-arylindolines containing a thiazole moiety were developed and found to be promising modulators of the programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) axis. Compound A30 exhibited outstanding biochemical activity, with an IC50 of 11.2 nM in a homogeneous time-resolved fluorescence assay. In the cell-based assay, A30 significantly promoted IFN-γ secretion and rescued T-cell proliferation, which were inhibited by PD-1 activation. Furthermore, A30 showed favorable in vivo antitumor activity in a mouse 4T1 breast carcinoma model. Moreover, in mouse CT26 colon carcinoma models, A30 potently suppressed the growth of CT26/PD-L1 tumor but did not obviously affect the growth of CT26/vector tumor. The results of flow cytometry analysis indicated that A30 inhibited tumor growth by activating the immune microenvironment. We concluded that A30 is a new starting point for further development of PD-1/PD-L1 interaction inhibitors as antitumor agents.


Asunto(s)
Antineoplásicos/química , Antígeno B7-H1/metabolismo , Indoles/química , Receptor de Muerte Celular Programada 1/metabolismo , Tiazoles/química , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Sitios de Unión , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Indoles/metabolismo , Indoles/farmacología , Indoles/uso terapéutico , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Mapas de Interacción de Proteínas/efectos de los fármacos , Relación Estructura-Actividad
15.
Curr Opin Immunol ; 69: 47-55, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33676271

RESUMEN

Although multiple immune checkpoint inhibitors (ICI) have been identified and tested in the clinic, antibodies blocking the PD-1/PD-L1 axis have produced the greatest impact on cancer treatment. Many potential mechanisms of treatment failure have been proposed from pre-clinical animal and human translational studies. Pre-clinical studies and clinical trials are underway to better understand how resistance arises and to develop strategies that can circumvent these resistance mechanisms and sensitize patients to anti-PD1/PD-L1 to improve clinical outcomes.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Humanos , Neoplasias/inmunología
16.
Mol Med Rep ; 23(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33760188

RESUMEN

Programmed cell death protein­1 (PD­1)/programmed death protein ligand­1 (PD­L1) inhibitors for treatment of a various types of cancers have revolutionized cancer immunotherapy. However, PD­1/PD­L1 inhibitors are associated with a low response rate and are only effective on a small number of patients with cancer. Development of an anti­PD­1/PD­L1 sensitizer for improving response rate and effectiveness of immunotherapy is a challenge. The present study reviews the synergistic effects of PD­1/PD­L1 inhibitor with oncolytic virus, tumor vaccine, molecular targeted drugs, immunotherapy, chemotherapy, radiotherapy, intestinal flora and traditional Chinese medicine, to provide information for development of effective combination therapies.


Asunto(s)
Antígeno B7-H1/genética , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/genética , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico/inmunología , Inmunoterapia/tendencias , Neoplasias/genética , Neoplasias/inmunología , Viroterapia Oncolítica/tendencias , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología
17.
J Ethnopharmacol ; 274: 113978, 2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-33716082

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Triple-negative breast cancer (TNBC) is the most aggressive and the worst prognosis breast cancer with limited treatment options. Taraxacum mongolicum (also called dandelion) is a traditional Chinese medicine has been used to treat mastitis, breast abscess, and hyperplasia of mammary glands since ancient times. In modern pharmacological research, dandelion has been proven with anti-breast cancer activities. We previously reported that dandelion extract could induce apoptosis in TNBC cells. However, its anti-tumor effects and mechanisms in the tumor microenvironment have not yet been elucidated. AIM OF THE STUDY: Tumor-associated macrophages (TAMs) play an important role in regulating the interaction between tumor cells and the immune system. The present study aimed to investigate the effects and mechanisms of dandelion extract on TNBC cells under the microenvironment of TAMs, as well as its influence on the polarization of M2 macrophages. MATERIALS AND METHODS: M2 macrophages were induced by phorbol-12-myristate 13-acetate (PMA) and interleukin 4 (IL-4), and verified by flow cytometry, quantitative RT-PCR (qRT-PCR), Western blotting, and ELISA. MDA-MB-231 and MDA-MB-468 TNBC cells were co-cultured with the supernatant of M2 macrophage which providing the TAMs microenvironment. The antitumor activity of dandelion extract in TNBC cells was evaluated by MTT assay. The invasive and migratory capacity of TNBC cells was measured by transwell assays. The expression of protein and gene was assessed by Western blotting and qRT-PCR, respectively. RESULTS: TAMs microenvironment promoted the proliferation, migration, and invasion of TNBC cells. However, dandelion extract inhibited the malignant property of MDA-MB-231 and MDA-MB-468 cells induced by TAMs. Both of TAMs and IL-10 caused STAT3 activation and PD-L1 higher expression, the immunosuppressive molecules in TNBC cells, and this effect can be attenuated by IL-10 neutralizing antibody. Dandelion extract exerted inhibition on STAT3 and PD-L1 in TNBC cells under TAMs microenvironment. Furthermore, in M2 macrophages, dandelion extract remarkably promoted the expression of M1-like marker TNF-α, IL-8, and iNOS, but reduced M2-like marker IL-10, CD206, Arginase-1, and TGF-ß. CONCLUSION: Dandelion extract inhibited the proliferation, migration and invasion of TNBC cells in TAMs microenvironment through suppressing IL-10/STAT3/PD-L1 immunosuppressive signaling pathway. Furthermore, dandelion extract promoted the polarization of macrophages from M2 to M1 phenotype. Thus, our results indicated that dandelion may serve as a promising therapeutic strategy for TNBC by modulating tumor immune microenvironment.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Medicamentos Herbarios Chinos/farmacología , Interleucina-10/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Taraxacum/química , Neoplasias de la Mama Triple Negativas/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/química , Humanos , Interleucina-10/metabolismo , Macrófagos/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral , Macrófagos Asociados a Tumores/efectos de los fármacos
19.
Invest New Drugs ; 39(3): 829-835, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33415580

RESUMEN

Background Treatment of recurrent, unresectable granulosa cell tumor (GCT) of the ovary can be challenging. Given the rarity of the tumor, alternative therapies have been difficult to evaluate in large prospective clinical trials. Currently, to our knowledge, there are no reports of the use of immune checkpoint inhibitors in GCT patients. Here, we present a case series of GCT patients treated with pembrolizumab who were enrolled in a phase II basket trial in advanced, rare solid tumors (ClinicalTrials.gov: NCT02721732). Cases We identified 5 patients with recurrent GCT (4 adult and 1 juvenile type); they had an extensive history of systemic therapy at study enrollment (range, 3-10), with most regimens resulting in less than 12 months of disease control. Pembrolizumab was administered in these patients, as per trial protocol. Although there were no objective responses according to the irRECIST guidelines, 2 patients with adult-type GCT experienced disease control for ≥ 12 months (565 and 453 days). In one, pembrolizumab represented the longest duration of disease control compared to prior lines of systemic therapy (565 days vs. 13 months). In the other, pembrolizumab was the second longest systemic therapy associated with disease control (453 days vs. 22 months) compared to prior lines of therapy. In this patient, pembrolizumab was discontinued following withdrawal of consent. PD-L1 expression was not observed in any baseline tumor samples. Pembrolizumab was well tolerated, with no grade 3 or 4 treatment-related adverse events. Conclusions Although our results do not support the routine use of pembrolizumab monotherapy in unselected GCT patients, some patients with adult-type GCT may derive a clinical benefit, with a low risk of toxicity. Future studies should investigate the role of immunotherapy and predictors of clinical benefit in this patient population.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Tumor de Células de la Granulosa/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Femenino , Proteína Forkhead Box L2/genética , Tumor de Células de la Granulosa/genética , Tumor de Células de la Granulosa/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Linfocitos Infiltrantes de Tumor/inmunología , Persona de Mediana Edad , Mutación , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Neurofibromina 1/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/genética , Adulto Joven
20.
Cancer Immunol Immunother ; 70(1): 31-45, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32623477

RESUMEN

Dendritic cell (DC)-based vaccines are recognized as a promising immunotherapeutic strategy against cancer; however, the efficacy of immunotherapy with DCs is controlled via immune checkpoints, such as programmed death-ligand 1 (PD-L1). PD-L1 expressed on DC and tumor cells binds to programmed death-1 (PD-1) receptors on the activated T cells, which leads to the inhibition of cytotoxic T cells. Blocking of PD-L1 on DC may lead to improve the efficacy of DC therapy for cancer. Here we demonstrated that DC vaccination in combination with pomalidomide and programmed death-ligand 1 (PD-L1) blockade inhibited tumor growth of a multiple myeloma (MM) mouse model. DCs + pomalidomide with dexamethasone + PD-L1 blockade significantly inhibited immune immunosuppressive factors and promoted proportions of immune effector cells in the spleen and tumor microenvironment. Additionally, functional activities of cytotoxic T lymphocytes and NK cells in spleen were enhanced by DCs + pomalidomide with dexamethasone + PD-L1 blockade. Taken together, this study identifies a potential new therapeutic approach for the treatment of MM. These results also provide a foundation for the future development of immunotherapeutic modalities to inhibit tumor growth and restore immune function in MM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Células Dendríticas/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/terapia , Talidomida/análogos & derivados , Animales , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Quimioterapia Adyuvante/métodos , Evaluación Preclínica de Medicamentos/métodos , Femenino , Inmunosupresores/farmacología , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Mieloma Múltiple/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Talidomida/farmacología , Vacunación/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA