Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
BMC Immunol ; 25(1): 60, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39271997

RESUMEN

BACKGROUND: Immune checkpoint inhibitor rechallenge has emerged as a prominent study area in non-small cell lung cancer (NSCLC). ß-glucan was reported to reverse resistance to anti-PD-1/PD-L1 inhibitors by regulating the tumor microenvironment. In this self-initiated clinical trial (ChiCTR2100054796), NSCLC participants who have previously failed anti-PD-1 therapy received ß-glucan (500 mg, bid, d1-21), Envafolimab (300 mg, d1) and Endostar (210 mg, civ72h) every 3 weeks until disease progression or unacceptable toxicity. The clinical efficacy and adverse events were observed, while serum samples were collected for proteomic analysis. RESULTS: Twenty Three patients were enrolled from January 2022 to March 2023 (median age, 65 years; male, n = 18 [78.3%]; squamous NSCLC, n = 9 [39.1%]; mutant type, n = 13 [56.5%]). The overall response rate (ORR) was 21.7% and disease control rate (DCR) was 73.9%. Median progression-free survival (mPFS) and median overall survival (mOS) was 4.3 months [95% CI: 2.0-6.6] and 9.8 months [95% CI: 7.2-12.4], respectively. The mPFS between PD-L1 positive and negative subgroup has significant difference (6.3 months vs. 2.3 months, p = 0.002). Treatment-related adverse events (TRAEs) occurred in 52.2% of patients. The most common TRAEs were hypothyroidism (26.1%) and fatigue (26.1%). 2 (8.7%) grade 3 adverse events were reported. No adverse reaction related deaths have been observed. Proteomic analysis revealed that the levels of CASP-8, ARG1, MMP12, CD28 and CXCL5 correlated with resistance to the treatment while the levels of CD40-L and EGF related to the favorable response. CONCLUSION: ß-glucan combined with Envafolimab and Endostar has considerable efficacy and safety for immune rechallenge in metastatic NSCLC patients who failed of anti-PD-1 treatment previously, especially for PD-L1 positive patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , beta-Glucanos , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Masculino , Femenino , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Anciano , Persona de Mediana Edad , beta-Glucanos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Metástasis de la Neoplasia , Resultado del Tratamiento
2.
Cancer Immunol Immunother ; 73(6): 103, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38630135

RESUMEN

As one of the scheduled immunization vaccines worldwide, virtually all individuals have been vaccinated with BCG vaccine. In order to verify the hypothesis that delivering BCG high-affinity peptides to tumor areas could activate the existing BCG memory T cells to attack tumor, we firstly predicted the HLA-A*0201 high-affinity peptides of BCG Ag85A protein (KLIANNTRV, GLPVEYLQV), and then, A375 melanoma cells and HLA-A*0201 PBMCs (from PPD-positive adults) were added to co-incubated with the predicted peptides in vitro. We found that the predicted BCG high-affinity peptides could be directly loaded onto the surface of tumor cells, enhancing the tumor-killing efficacy of PBMCs from PPD-positive volunteer. Then, we constructed PPD-positive mice model bearing B16F10 subcutaneous tumors and found that intratumor injection of BCG Ag85A high-affinity peptides (SGGANSPAL, YHPQQFVYAGAMSGLLD) enhanced the anti-tumor efficacy in PPD-positive melanoma mice. Along with the better anti-tumor efficacy, the expression of PDL1 on tumor cell surface was also increased, and stronger antitumor effects occurred when further combined with anti-PD1 antibody. For microenvironment analysis, the proportion of effector memory T cells was increased and the better treatment efficacy may be attributed to the elevated effector memory CD4 + T cells within the tumor. In conclusion, using the existing immune response of BCG vaccine by delivering high-affinity peptides of BCG to tumor area is a safe and promising therapy for cancer.


Asunto(s)
Melanoma , Humanos , Adulto , Animales , Ratones , Melanoma/tratamiento farmacológico , Vacuna BCG/uso terapéutico , Péptidos , Modelos Animales de Enfermedad , Inmunización , Microambiente Tumoral
3.
J Transl Med ; 21(1): 619, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37700338

RESUMEN

BACKGROUND: In situ tumor vaccine has been gradually becoming a hot research field for its advantage of achieving personalized tumor therapy without prior antigen identification. Various in situ tumor vaccine regimens have been reported to exert considerable antitumor efficacy in preclinical and clinical studies. However, the design of in situ tumor vaccines still needs further optimization and the underlying immune mechanism also waits for deeper investigation. METHODS: A novel triple in situ vaccine strategy that combining local radiation with intratumoral injection of TLR9 agonist CpG and OX40 agonist was established in this sturdy. Local and abscopal antitumor efficacy as well as survival benefit were evaluated in the bilateral tumors and pulmonary metastasis model of B16F10 melanoma. In situ vaccine-induced immune responses and immune-associated variation in tumor environment were further investigated using multiparameter flow cytometry and RNA sequencing. Base on the analysis, the RT + CpG + αOX40 triple in situ vaccine was combined with checkpoint blockade therapy to explore the potential synergistic antitumor efficacy. RESULTS: Enhanced tumor suppression was observed with minimal toxicity in both treated and untreated abscopal tumors after receiving RT + CpG + αOX40 triple vaccine. The introduction of local radiation and OX40 agonist benefit more to the inhibition of local and abscopal lesions respectively, which might be partially attributed to the increase of effector memory T cells in the tumor microenvironment. Further analysis implied that the triple in situ vaccine did not only activate the microenvironment of treated tumors, with the upregulation of multiple immune-associated pathways, but also enhanced systemic antitumor responses, thus achieved superior systemic tumor control and survival benefit. Moreover, the triple in situ vaccine synergized with checkpoint blockade therapy, and significantly improved the therapeutic effect of anti-programmed cell death protein (PD)-1 antibody. CONCLUSION: This triple combining in situ vaccine induced intensive antitumor responses, mediated effective systemic tumor control and survival benefit, and displayed impressive synergistic antitumor effect with checkpoint blockade therapy. These data preliminary confirmed the efficacy, feasibility and safety of the triple combining in situ vaccine, suggesting its great application potential as both monotherapy and a part of combined immunotherapeutic regimens in clinical scenario.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Humanos , Vacunas contra el Cáncer/uso terapéutico , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Anticuerpos , Citometría de Flujo , Microambiente Tumoral
4.
J Transl Med ; 21(1): 78, 2023 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-36739402

RESUMEN

BACKGROUND: Acral melanoma (AM) is the most common subtype in Chinese melanoma patients with a very poor prognosis. However, our understanding of the disease pathogenesis and molecular landscape is limited by the few studies that have been conducted. Here, we profiled the clinical characteristics, mutational landscapes and tumor immune microenvironment of AM patients to gain insights into disease characteristics and potential treatment strategies. METHODS: A total of 90 AM patients were enrolled and their tissue samples were subjected to next-generation sequencing and multiplexed immunohistochemistry tests. Kaplan-Meier curves and log-rank tests were used to analyze the prognostic potential of various genetic aberrations and immune cell compositions in AM. RESULTS: The median disease-free survival was 21.3 months and estimated median overall survival (OS) was 60 months. More advanced stages, older ages and thickness of greater than 4 mm were associated with worse prognosis in AM patients (HR = 2.57, 95% CI 1.25-5.29, p = 0.01; HR = 2.77, 95% CI 1.22-6.28, p = 0.02; HR = 3.43, 95% CI 1.51-7.82, p < 0.01, respectively), while patients who received post-surgical treatments had better survival (HR = 0.36, 95% CI 0.17-0.76, p = 0.01). The most frequently altered genes included BRAF (14.5%), KIT (16.9%), NRAS (12%), NF1 (10.8%), APC (7.2%), and ARID2 (6%). Copy number variations (CNV) were commonly found in CCND1 (19.3%), CDK4 (19.3%), MDM2 (14.5%) and FGF19 (12%). CDK4 amplifications was independently associated with shorter OS in AM patients (HR = 3.61, 95% CI 1.38-9.46, p = 0.01). CD8 + T cells (p < 0.001) and M1 macrophages (p = 0.05) were more highly enriched in the invasive margin than in the tumor center. Patients with higher levels of M1 macrophage infiltration in the invasive margin derived markedly longer OS (HR = 0.43, 95% CI 0.20-0.95, p = 0.03). Interestingly, in CDK4-amplified patients, there tended to be a low level of M1 macrophage infiltration in the invasive margin (p = 0.06), which likely explains the poor prognosis in such patients. CONCLUSIONS: Our study provided a comprehensive portrait of the clinicopathological features, genetic aberrations and tumor microenvironment profiles in AM patients and identified candidate prognostic factors, which may facilitate development of additional therapeutic options and better inform clinical management of AM patients. Based on these prognostic factors, further studies should focus on enhancing the infiltration of M1 macrophages, especially in CDK4-amplified AM patients.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Pronóstico , Variaciones en el Número de Copia de ADN/genética , Microambiente Tumoral/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Melanoma Cutáneo Maligno
5.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-961954

RESUMEN

@#[摘 要] 目的:探讨肝转移对晚期胃癌患者免疫治疗效果的影响。方法:收集2019年2月至2022年1月在南京医科大学附属常州第二人民医院肿瘤中心接受过免疫治疗的晚期胃癌患者的临床资料,进行回顾性分析,利用卡方检验或Fisher确切概率法进行基线特征比较,利用卡方检验和Kaplan-Meier生存分析方法进行有肝转移与无肝转移胃癌患者的疗效和生存期的比较。结果:共有48例晚期胃癌患者纳入分析,根据有无肝转移将患者分为肝转移队列(n=20)和无肝转移队列(n=28)。有肝转移较无肝转移胃癌患者体力状况更差。肝转移队列与无肝转移队列的ORR分别为15.0%和35.7%(P>0.05),DCR分别为65.0%和82.1%(P>0.05);中位PFS在两组分别为5.0个月和11.2个月(HR=0.40,P<0.05),中位OS分别为12.0个月和19.0个月(P>0.05)。结论:胃癌肝转移患者免疫治疗的疗效差于无肝转移的患者。

6.
Chin J Integr Med ; 28(10): 909-917, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34913148

RESUMEN

OBJECTIVE: To verrify the anti-tumor efficacy and toxicity between juglone (Jug) and Jug-loaded PLGA nanoparticles (Jug-PLGA-NPs). METHODS: Jug-PLGA-NPs were prepared by ultrasonic emulsification. The anti-tumor activity of Jug (2, 3, 4 µg/mL) and Jug-PLGA-NPs (Jug: 2, 3, 4 µg/mL) in vitro was measured by MTT assay and cell apoptosis analysis. The distribution, anti-tumor effect and biological safety in vivo was evaluated on A375 nude mice. RESULTS: With the advantage of good penetration and targeting properties, Jug-PLGA-NPs significantly inhibited proliferation and migration of melanoma cells both in vitro and in vivo (P<0.05 or P<0.01) with acceptable biocompatibility. CONCLUSIONS: Jug can inhibit the growth of melanoma but is highly toxic. With the advantage of sustained release, tumor targeting, anti-tumor activity and acceptable biological safety, Jug-PLGA-NPs provide a new pharmaceutical form for future application of Jug.


Asunto(s)
Melanoma , Nanopartículas , Animales , Línea Celular Tumoral , Preparaciones de Acción Retardada/uso terapéutico , Portadores de Fármacos/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Ratones Desnudos , Naftoquinonas , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico
7.
Front Oncol ; 11: 692480, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745936

RESUMEN

A 48-year old woman was diagnosed with metastatic pancreatic acinar cell carcinoma (PACC) and with a marked elevation in alpha-fetoprotein (AFP), this being a recognized but uncommon feature of PACC. As she refused chemotherapy, the combined therapy of lenvatinib and sintilimab (lenvatinib 8 mg, orally, qd; and sintilimab 100 mg, intravenous glucose tolerance test, q21d) was given, which conferred significant tumor shrinkage and long progression-free survival (>21 months). This study is the first report and description of a PACC demonstrating favorable response to the combination therapy of an antiangiogenic agent and immunotherapy.

8.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-886479

RESUMEN

@#[摘 要] 目的:探讨恶性黑色素瘤(malignant melanoma,MM)微环境分型对MM患者预后的评估价值。方法:对2010年7月至2017年5月在南京鼓楼医院手术切除的87例原发性MM组织进行二代测序,免疫组化法检测PD-1、PD-L1、CD3+ TIL、MSH2、MSH6、PMS2和MLH1的表达。随访患者的生存时间,分析不同免疫微环境分型对患者预后的影响及其基因表达特征。结果:根据PD-L1和TIL表达水平将87例MM患者的肿瘤微环境分为4个亚型:PD-L1+ TIL+型或双阳型(15/87,17.24%)、PD-L1+ TIL-型(15/87,17.24%)、PD-L1- TIL+型(20/87,22.99%)、PD-L1- TIL-型或双阴型(37/87,42.53%)。双阳型患者的中位无病生存期显著长于双阴型患者(P<0.05),此可能与双阴型患者存在更多CDK4、MCL1、MYC、AKT2、CCND1、FGF19等预后不良基因拷贝数扩增相关;双阳型患者PD-1表达显著高于双阴型患者(P<0.01),可能与PD-L1、TIL分别与PD-1呈共表达和共不表达有关。结论:根据PD-L1及TIL表达将MM 患者微环境分为4种亚型,能够区分MM患者预后,双阴型患者存在更多预后不良基因拷贝数扩增。

9.
Pigment Cell Melanoma Res ; 33(4): 601-611, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31944535

RESUMEN

The incidence of melanoma is rising globally including China. Comparing to Caucasians, the incidence of non-cutaneous melanomas is significantly higher in Chinese. Herein, we performed genomic profiling of 89 Chinese surgically resected primary melanomas, including acral (n = 54), cutaneous (n = 22), and mucosal (n = 13), by hybrid capture-based next-generation sequencing. We show that mucosal melanomas tended to harbor more pathogenic mutations than other types of melanoma, though the biological significance of this finding remains uncertain. Chromosomal arm-level alterations including 6q, 9p, and 10p/q loss were highly recurrent in all subtypes, but mucosal melanoma was significantly associated with increased genomic instability. Importantly, 7p gain significantly correlated with unfavorable clinical outcomes in non-cutaneous melanomas, representing an intriguing prognostic biomarker of those subtypes. Furthermore, focal amplification of 4q12 (KIT, KDR, and PDGFRα) and RAD51 deletion were more abundant in mucosal melanoma, while NOTCH2 amplification was enriched in acral melanoma. Additionally, cutaneous melanomas had higher mutation load than acral melanomas, while mucosal melanomas did not differ from other subtypes in mutation burden. Together, our data revealed important features of acral and mucosal melanomas in Chinese including distinctive driver mutation pattern and increased genomic instability. These findings highlight the possibilities of combination therapies in the clinical management of melanoma.


Asunto(s)
Análisis Mutacional de ADN , Genómica , Melanoma/genética , Membrana Mucosa/patología , Mutación/genética , Adulto , Anciano , Anciano de 80 o más Años , Pueblo Asiatico/genética , Supervivencia sin Enfermedad , Etnicidad/genética , Femenino , Dosificación de Gen , Humanos , Estimación de Kaplan-Meier , Masculino , Melanoma/enzimología , Persona de Mediana Edad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA