chemoimmunotherapy

化学免疫疗法
  • 文章类型: Journal Article
    化学免疫疗法是临床上治疗几种恶性疾病的新兴范例。如非小细胞肺癌,乳腺癌,和大B细胞淋巴瘤.然而,该策略的有效性仍然受到严重不良事件和高治疗终止率的限制,可能是由于缺乏化疗和免疫治疗剂的肿瘤靶向分布。靶向药物递送具有解决该问题的潜力。在临床转化中最有前途的纳米载体中,近年来,脂质体在肿瘤化疗免疫治疗中引起了广泛关注。脂质体支持的癌症化学免疫疗法在临床上取得了重大进展,令人印象深刻的治疗结果。本文综述了脂质体介导的癌症化学免疫治疗的最新临床前和临床进展,并讨论了该领域面临的挑战和未来的发展方向。
    Chemoimmunotherapy is an emerging paradigm in the clinic for treating several malignant diseases, such as non-small cell lung cancer, breast cancer, and large B-cell lymphoma. However, the efficacy of this strategy is still restricted by serious adverse events and a high therapeutic termination rate, presumably due to the lack of tumor-targeted distribution of both chemotherapeutic and immunotherapeutic agents. Targeted drug delivery has the potential to address this issue. Among the most promising nanocarriers in clinical translation, liposomes have drawn great attention in cancer chemoimmunotherapy in recent years. Liposomes-enabled cancer chemoimmunotherapy has made significant progress in clinics, with impressive therapeutic outcomes. This review summarizes the latest preclinical and clinical progress in liposome-enabled cancer chemoimmunotherapy and discusses the challenges and future directions of this field.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    耐药性是癌症化疗中的重大挑战,并且是导致癌症患者恢复不良的主要因素。尽管载药纳米颗粒在克服化疗耐药性方面显示出了希望,他们经常携带药物组合,需要先进的设计和制造工艺。此外,从免疫疗法的角度来看,他们很少接近化疗耐药的肿瘤。在这项研究中,我们开发了一种仅由化疗诱导的耐药肿瘤抗原(CIRTAs)和免疫佐剂Toll样受体(TLR)7/8激动剂R848(CIRTAs@R848)组成的治疗性纳米疫苗.这种纳米疫苗不需要额外的载体并且具有简单的生产过程。它有效地同时向树突状细胞(DCs)递送抗原和免疫刺激剂,促进DC成熟。CIRTAs@R848表现出显著的肿瘤抑制,特别是与免疫检查点阻断(ICB)抗PD-1(αPD-1)联合使用时。联合治疗增加了T细胞向肿瘤的浸润,同时降低了调节性T细胞(Tregs)的比例并调节了肿瘤微环境,导致长期免疫记忆。总的来说,这项研究从一个新的角度介绍了一种治疗化疗耐药肿瘤的创新策略,具有在个性化免疫疗法和精准医学中的潜在应用。
    Drug resistance is a significant challenge in cancer chemotherapy and is a primary factor contributing to poor recovery for cancer patients. Although drug-loaded nanoparticles have shown promise in overcoming chemotherapy resistance, they often carry a combination of drugs and require advanced design and manufacturing processes. Furthermore, they seldom approach chemotherapy-resistant tumors from an immunotherapy perspective. In this study, we developed a therapeutic nanovaccine composed solely of chemotherapy-induced resistant tumor antigens (CIRTAs) and the immune adjuvant Toll-like receptor (TLR) 7/8 agonist R848 (CIRTAs@R848). This nanovaccine does not require additional carriers and has a simple production process. It efficiently delivers antigens and immune stimulants to dendritic cells (DCs) simultaneously, promoting DCs maturation. CIRTAs@R848 demonstrated significant tumor suppression, particularly when used in combination with the immune checkpoint blockade (ICB) anti-PD-1 (αPD-1). The combined therapy increased the infiltration of T cells into the tumor while decreasing the proportion of regulatory T cells (Tregs) and modulating the tumor microenvironment, resulting in long-term immune memory. Overall, this study introduces an innovative strategy for treating chemotherapy-resistant tumors from a novel perspective, with potential applications in personalized immunotherapy and precision medicine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    采用靶向内质网(ER)的化学免疫疗法的前景为放大化学疗法和免疫疗法的协同作用提供了机会。在这项研究中,我们最初通过促进结直肠癌(CRC)细胞内质网应激和自噬,验证了雷公藤红素(CEL)作为免疫原性细胞死亡(ICD)的诱导剂.随后,提出了以ER为目标的策略,涉及使用KDEL肽修饰的来自牛奶的外泌体(KME)将CEL与PD-L1小干扰RNA(siRNA)共同递送,以增强化学免疫疗法的结果。我们的发现证明了KME通过高尔基体到ER途径的有效运输。与他们的非目标对手相比,KME表现出CEL诱导的ICD效应的显著增强。此外,它促进了危险信号分子(DAMPs)的释放,从而刺激树突状细胞的抗原呈递功能并促进T细胞向肿瘤的浸润。同时,ER靶向递送PD-L1siRNA导致细胞内和膜PD-L1蛋白表达下调,从而促进CD8+T细胞的增殖和活性。最终,ER靶向制剂表现出增强的抗肿瘤功效,并在体内引发针对原位结直肠肿瘤的抗肿瘤免疫应答.总的来说,稳健的ER靶向递送策略为实现有效的癌症化学免疫疗法提供了令人鼓舞的方法.
    The prospect of employing chemoimmunotherapy targeted towards the endoplasmic reticulum (ER) presents an opportunity to amplify the synergistic effects of chemotherapy and immunotherapy. In this study, we initially validated celastrol (CEL) as an inducer of immunogenic cell death (ICD) by promoting ER stress and autophagy in colorectal cancer (CRC) cells. Subsequently, an ER-targeted strategy was posited, involving the codelivery of CEL with PD-L1 small interfering RNAs (siRNA) using KDEL peptide-modified exosomes derived from milk (KME), to enhance chemoimmunotherapy outcomes. Our findings demonstrate the efficient transportation of KME to the ER via the Golgi-to-ER pathway. Compared to their non-targeting counterparts, KME exhibited a significant augmentation of the CEL-induced ICD effect. Additionally, it facilitated the release of danger signaling molecules (DAMPs), thereby stimulating the antigen-presenting function of dendritic cells and promoting the infiltration of T cells into the tumor. Concurrently, the ER-targeted delivery of PD-L1 siRNA resulted in the downregulation of both intracellular and membrane PD-L1 protein expression, consequently fostering the proliferation and activity of CD8+ T cells. Ultimately, the ER-targeted formulation exhibited enhanced anti-tumor efficacy and provoked anti-tumor immune responses against orthotopic colorectal tumors in vivo. Collectively, a robust ER-targeted delivery strategy provides an encouraging approach for achieving potent cancer chemoimmunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:非小细胞肺癌(NSCLC)是肺癌的主要形式,化疗和免疫治疗的结合为患有这种疾病的患者提供了有希望的治疗选择。然而,耐药性的出现显著限制了这些治疗策略的有效性.因此,必须设计出准确检测和评估这些治疗方法的疗效的方法。
    目的:确定非小细胞肺癌患者中与中性粒细胞胞外陷阱(NETs)和化学免疫疗法疗效相关的代谢特征。
    方法:总共,纳入159例接受一线化学免疫治疗的NSCLC患者。我们首先调查了影响临床疗效的特征。通过商业试剂盒测量NETs和细胞因子的循环水平。液相色谱串联质谱定量血浆代谢物,并鉴定了差异代谢物。最小绝对收缩和选择运算符,支持向量机-递归特征消除,并采用随机森林算法。通过使用血浆代谢谱和机器学习算法,建立了预测性代谢特征。
    结果:首先,循环白细胞介素-8的水平,中性粒细胞与淋巴细胞的比率,和NETs与一线化学免疫疗法疗效差密切相关。将患者分为低NET组或高NET组。总共鉴定了54种差异血浆代谢物。这些代谢物主要参与花生四烯酸和嘌呤代谢。三个关键代谢物被确定为关键变量,包括8,9-环氧二十碳三烯酸,L-苹果酸,和双(单酰基甘油)磷酸酯(18:1/16:0)。使用代谢组学测序数据和机器学习方法,筛选关键代谢特征以预测NET水平和化学免疫疗法疗效.
    结论:确定的代谢特征可以有效区分NET水平,并预测NSCLC患者化学免疫疗法的临床获益。
    BACKGROUND: Non-small cell lung cancer (NSCLC) is the primary form of lung cancer, and the combination of chemotherapy with immunotherapy offers promising treatment options for patients suffering from this disease. However, the emergence of drug resistance significantly limits the effectiveness of these therapeutic strategies. Consequently, it is imperative to devise methods for accurately detecting and evaluating the efficacy of these treatments.
    OBJECTIVE: To identify the metabolic signatures associated with neutrophil extracellular traps (NETs) and chemoimmunotherapy efficacy in NSCLC patients.
    METHODS: In total, 159 NSCLC patients undergoing first-line chemoimmunotherapy were enrolled. We first investigated the characteristics influencing clinical efficacy. Circulating levels of NETs and cytokines were measured by commercial kits. Liquid chromatography tandem mass spectrometry quantified plasma metabolites, and differential metabolites were identified. Least absolute shrinkage and selection operator, support vector machine-recursive feature elimination, and random forest algorithms were employed. By using plasma metabolic profiles and machine learning algorithms, predictive metabolic signatures were established.
    RESULTS: First, the levels of circulating interleukin-8, neutrophil-to-lymphocyte ratio, and NETs were closely related to poor efficacy of first-line chemoimmunotherapy. Patients were classed into a low NET group or a high NET group. A total of 54 differential plasma metabolites were identified. These metabolites were primarily involved in arachidonic acid and purine metabolism. Three key metabolites were identified as crucial variables, including 8,9-epoxyeicosatrienoic acid, L-malate, and bis(monoacylglycerol)phosphate (18:1/16:0). Using metabolomic sequencing data and machine learning methods, key metabolic signatures were screened to predict NET level as well as chemoimmunotherapy efficacy.
    CONCLUSIONS: The identified metabolic signatures may effectively distinguish NET levels and predict clinical benefit from chemoimmunotherapy in NSCLC patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    以铂(II)为基础的药物(PtII),阻碍DNA复制,是最广泛使用的化疗药物。然而,目前的PtII药物经常错过它们的DNA靶标,导致严重的副作用和耐药性。为了克服这一挑战,我们开发了一种新型的奥沙利铂(IV)(PtIV)前药两亲物(C16-OPtIV-R8K),整合长链疏水脂质和靶向核的亲水肽(R8K)。这种设计允许前药自组装成高度均匀的脂质纳米颗粒(NTPtIV),用于增强靶向化疗和免疫疗法。随后,NTPtIV的生物活性和效应在不同的水平上进行了检查,包括癌细胞,3D肿瘤球,和体内。我们的体外研究表明,74%的铂类药物的癌细胞核定位比奥沙利铂高3.6倍,在消除耐药癌细胞方面实现了十倍以上的增长。在体内,NTPtIV显示有效的肿瘤积累,导致小鼠乳腺癌的肿瘤生长受到抑制。此外,NTPtIV招募更多的CD4+和CD8+T细胞,减少CD4+Foxp3+Tregs,协同增强靶向化疗和免疫治疗。总的来说,这种策略在核靶向癌症治疗中取得了有希望的进展,协同提高化疗和免疫治疗的疗效。
    Platinum(II)-based drugs (PtII), which hinder DNA replication, are the most widely used chemotherapeutics. However, current PtII drugs often miss their DNA targets, leading to severe side effects and drug resistance. To overcome this challenge, we developed a oxaliplatin-based platinum(IV) (PtIV) prodrug amphiphile (C16-OPtIV-R8K), integrating a long-chain hydrophobic lipid and a nucleus-targeting hydrophilic peptide (R8K). This design allows the prodrug to self-assemble into highly uniform lipid nanoparticles (NTPtIV) for enhanced targeting chemotherapy and immunotherapy. Subsequently, NTPtIV\'s bioactivity and effects were examined at diverse levels, encompassing cancer cells, 3D tumor spheres, and in vivo. Our in vitro studies show a 74% cancer cell nucleus localization of platinum drugs-3.6 times higher than that of oxaliplatin, achieving more than a ten-fold increase in eliminating drug-resistant cancer cells. In vivo, NTPtIV shows efficient tumor accumulation, leading to suppressed tumor growth of murine breast cancer. Moreover, NTPtIV recruited more CD4+ and CD8+ T cells and reduced CD4+ Foxp3+ Tregs to synergistically enhance targeted chemotherapy and immunotherapy. Overall, this strategy presents a promising advancement in nucleus-targeted cancer therapy, synergistically boosting the efficacy of chemotherapy and immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:对于可切除的非小细胞肺癌(NSCLC),推荐新辅助化学免疫疗法和手术治疗。然而,相当比例的患者没有接受手术,而是选择替代治疗,如放疗。放疗在这种情况下的疗效尚不清楚。
    方法:这项回顾性研究分析了III期NSCLC患者的数据,这些患者接受了新辅助化学免疫疗法以及手术或放疗。使用倾向评分匹配(PSM)来平衡组间的异质性。疗效结果,安全概况,并评估疾病复发模式。
    结果:总计,纳入175例患者;50例接受放疗,125人接受了手术。在匹配之前,在无进展生存期方面,放疗不如手术(PFS;危险比[HR],2.23;P=0.008)。在1:1PSM调整后,每组40例患者.放疗组中位PFS为30.8个月,手术组未达到(HR,1.46;P=0.390)。放疗组12个月和24个月的PFS分别为90.4%和69.0%,手术组为94.1%和73.9%,分别。PSM后的亚组分析表明,IIIA期疾病的患者比IIIB期疾病的患者从手术中受益更多(HR,3.00;P=0.074)。3-4级治疗相关不良事件(TRAEs)发生在放疗组的62.5%和手术组的55.0%,没有5级TRAE报告。放疗组和手术组3-4级治疗相关性肺炎或肺炎的发生率分别为7.5%和2.5%,分别。
    结论:对于初次新辅助化学免疫疗法后未接受手术切除的可切除非小细胞肺癌患者,放射治疗可能是一种可行的替代手术方法。提供相当的疗效和可控的安全性。需要更大的前瞻性研究来验证这些发现并优化该患者人群的治疗策略。
    BACKGROUND: Neoadjuvant chemoimmunotherapy followed by surgery is recommended for resectable non-small-cell lung cancer (NSCLC). However, a considerable proportion of patients do not undergo surgery and opt for alternative treatments such as radiotherapy. The efficacy of radiotherapy in this context remains unclear.
    METHODS: This retrospective study analyzed data from patients with stage III NSCLC who received neoadjuvant chemoimmunotherapy followed by either surgery or radiotherapy. Propensity score matching (PSM) was used to balance the heterogeneity between the groups. Efficacy outcomes, safety profiles, and disease recurrence patterns were assessed.
    RESULTS: In total, 175 patients were included; 50 underwent radiotherapy, and 125 underwent surgery. Prior to matching, radiotherapy was inferior to surgery in terms of progression-free survival (PFS; Hazard ratio [HR], 2.23; P = 0.008). Following a 1:1 PSM adjustment, each group consisted of 40 patients. The median PFS was 30.8 months in the radiotherapy group and not reached in the surgery group (HR, 1.46; P = 0.390). The 12- and 24-month PFS rates were 90.4 % and 69.0 % for the radiotherapy group compared to 94.1 % and 73.9 % for the surgery group, respectively. Subgroup analyses after PSM showed that patients with stage IIIA disease tend to benefit more from surgery than those with stage IIIB disease (HR, 3.00; P = 0.074). Grade 3-4 treatment-related adverse events (TRAEs) occurred in 62.5 % of patients in the radiotherapy group and 55.0 % in the surgery group, with no grade 5 TRAEs reported. The incidence of grade 3-4 treatment-related pneumonitis or pneumonia was 7.5 % and 2.5 % in the radiotherapy and surgery groups, respectively.
    CONCLUSIONS: Radiotherapy may be a viable alternative to surgery in patients with resectable NSCLC who do not undergo surgical resection after initial neoadjuvant chemoimmunotherapy, offering comparable efficacy and a manageable safety profile. Larger prospective studies are needed to validate these findings and optimize the treatment strategies for this patient population.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    本研究旨在建立基于肺功能检查的综合临床预后风险模型。该模型旨在指导接受新辅助化学免疫疗法的可切除I-III期非小细胞肺癌(NSCLC)患者的评估和预测管理。
    收集175例患者的临床病理特征和预后生存数据。单变量和多变量Cox回归分析,采用最小绝对收缩和选择算子(LASSO)回归分析来识别变量并构建相应的模型。整合这些变量以建立岭回归模型。对模型的鉴别和校准进行了评估,并在内部验证后选择了最佳模型。将最优模型的风险评分或分组与临床因素进行比较分析,以探讨其潜在的临床应用价值。
    单变量回归分析确定吸烟,完全病理反应(CPR),和主要病理反应(MPR)作为保护因素。相反,T分期,D-二聚体/白细胞比值(DWBCR),D-二聚体/纤维蛋白原比值(DFR),D-二聚体/分钟通气量实际比值(DMVAR)为危险因素。对这些模型的评估证实了它们准确预测患者预后的能力,表现出理想的辨别和校准,岭回归模型是最优的。生存分析表明,高危组(HRG)的无病生存期(DFS)明显短于低危组(LRG)(P=2.57×10-13)。时间依赖性受试者工作特征(ROC)曲线表明,1年时的曲线下面积(AUC)值,2年,3年分别为0.74、0.81和0.79。临床相关分析显示,男性肺鳞状细胞癌或慢性阻塞性肺疾病(COPD)患者以LRG为主,提示更好的预后,并可能确定该治疗组合的受益人群。
    本研究开发的预后模型可有效预测接受新辅助化学免疫治疗的非小细胞肺癌患者的预后。它为临床医生提供了有价值的预测性见解,协助制定治疗计划和监测疾病进展。
    UNASSIGNED: This study aimed to establish a comprehensive clinical prognostic risk model based on pulmonary function tests. This model was intended to guide the evaluation and predictive management of patients with resectable stage I-III non-small cell lung cancer (NSCLC) receiving neoadjuvant chemoimmunotherapy.
    UNASSIGNED: Clinical pathological characteristics and prognostic survival data for 175 patients were collected. Univariate and multivariate Cox regression analyses, and least absolute shrinkage and selection operator (LASSO) regression analysis were employed to identify variables and construct corresponding models. These variables were integrated to develop a ridge regression model. The models\' discrimination and calibration were evaluated, and the optimal model was chosen following internal validation. Comparative analyses between the risk scores or groups of the optimal model and clinical factors were conducted to explore the potential clinical application value.
    UNASSIGNED: Univariate regression analysis identified smoking, complete pathologic response (CPR), and major pathologic response (MPR) as protective factors. Conversely, T staging, D-dimer/white blood cell ratio (DWBCR), D-dimer/fibrinogen ratio (DFR), and D-dimer/minute ventilation volume actual ratio (DMVAR) emerged as risk factors. Evaluation of the models confirmed their capability to accurately predict patient prognosis, exhibiting ideal discrimination and calibration, with the ridge regression model being optimal. Survival analysis demonstrated that the disease-free survival (DFS) in the high-risk group (HRG) was significantly shorter than in the low-risk group (LRG) (P=2.57×10-13). The time-dependent receiver operating characteristic (ROC) curve indicated that the area under the curve (AUC) values at 1 year, 2 years, and 3 years were 0.74, 0.81, and 0.79, respectively. Clinical correlation analysis revealed that men with lung squamous cell carcinoma or comorbid chronic obstructive pulmonary disease (COPD) were predominantly in the LRG, suggesting a better prognosis and potentially identifying a beneficiary population for this treatment combination.
    UNASSIGNED: The prognostic model developed in this study effectively predicts the prognosis of patients with NSCLC receiving neoadjuvant chemoimmunotherapy. It offers valuable predictive insights for clinicians, aiding in developing treatment plans and monitoring disease progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    将免疫疗法与基于纳米材料的化学疗法整合为放大抗肿瘤结果提供了有希望的途径。然而,化疗引起的抑制性肿瘤免疫微环境(TIME)和环氧合酶-2(COX-2)上调可能会阻碍化学免疫治疗的疗效。这项研究通过开发空间位阻效应调节的锌基金属有机框架(MOF)提出了一种时间重塑策略,指定为CZFNP。这种纳米反应器是通过原位加载COX-2抑制剂而设计的,C-藻蓝蛋白(CPC),进入框架构建块,同时削弱了MOF的稳定性。因此,CZFNP在特异性转运至过度表达叶酸受体的肿瘤细胞时实现锌离子(Zn2+)和CPC的快速pH响应性释放。因此,Zn2+可以诱导活性氧(ROS)介导的细胞毒性治疗,同时与线粒体DNA(mtDNA)释放同步,刺激mtDNA/cGAS-STING通路介导的先天免疫。CPC抑制化疗诱导的COX-2过表达,从而共同重新编程抑制性TIME并增强抗肿瘤免疫反应。在异种移植肿瘤模型中,CZFNPs系统有效调节STING和COX-2表达,将“冷”肿瘤转化为“热”肿瘤,从而导致相对于单独的ZIF-8治疗约4倍的肿瘤消退。这种方法为增强基于纳米材料的联合化疗和免疫疗法的功效提供了有效的策略。
    Integrating immunotherapy with nanomaterials-based chemotherapy presents a promising avenue for amplifying antitumor outcomes. Nevertheless, the suppressive tumor immune microenvironment (TIME) and the upregulation of cyclooxygenase-2 (COX-2) induced by chemotherapy can hinder the efficacy of the chemoimmunotherapy. This study presents a TIME-reshaping strategy by developing a steric-hindrance effect tuned zinc-based metal-organic framework (MOF), designated as CZFNPs. This nanoreactor is engineered by in situ loading of the COX-2 inhibitor, C-phycocyanin (CPC), into the framework building blocks, while simultaneously weakening the stability of the MOF. Consequently, CZFNPs achieve rapid pH-responsive release of zinc ions (Zn2+) and CPC upon specific transport to tumor cells overexpressing folate receptors. Accordingly, Zn2+ can induce reactive oxygen species (ROS)-mediated cytotoxicity therapy while synchronize with mitochondrial DNA (mtDNA) release, which stimulates mtDNA/cGAS-STING pathway-mediated innate immunity. The CPC suppresses the chemotherapy-induced overexpression of COX-2, thus cooperatively reprogramming the suppressive TIME and boosting the antitumor immune response. In xenograft tumor models, the CZFNPs system effectively modulates STING and COX-2 expression, converting \"cold\" tumors into \"hot\" tumors, thereby resulting in ≈ 4-fold tumor regression relative to ZIF-8 treatment alone. This approach offers a potent strategy for enhancing the efficacy of combined nanomaterial-based chemotherapy and immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    干扰素调节因子1(IRF1)在非小细胞肺癌(NSCLC)化疗中的临床应用及生物学功能研究尚不清楚。这项研究的目的是探讨IRF1在NSCLC患者中的预测和预后意义。我们使用cBioPortal数据库来预测IRF1的频率变化并探索其靶基因。采用生物信息学方法分析IRF1与免疫调节因子的关系。对临床样本进行回顾性分析,以评估IRF1在化学免疫治疗中的预测和预后价值。此外,构建具有不同IRF1表达水平的A549细胞以研究其对NSCLC细胞的影响。同时进行动物实验以研究IRF1在体内的作用。我们的发现表明IRF1的主要突变是深度缺失,并且与免疫调节因子密切相关。KRAS和TP53是IRF1的靶基因之一,其中干扰素和IL-2是主要受影响的途径。临床上,IRF1水平与化学免疫疗法的疗效显着相关。IRF1水平高的患者的中位无进展生存期(mPFS)为9.5个月,而IRF1水平低的患者的mPFS较短,为5.8个月.IRF1水平与PD-L1分布和循环IL-2水平呈正相关。IL-2增强了IRF1的生物学功能,并概括了其在敲除组中的体内作用。因此,IRF1可能通过调节IL-2炎症途径对NSCLC患者的化学免疫治疗具有预测和预后价值。
    The clinical application and biological function of interferon regulatory factor 1 (IRF1) in non-small cell lung cancer (NSCLC) patients undergoing chemoimmunotherapy remain elusive. The aim of this study was to investigate the predictive and prognostic significance of IRF1 in NSCLC patients. We employed the cBioPortal database to predict frequency changes in IRF1 and explore its target genes. Bioinformatic methods were utilized to analyze the relationship between IRF1 and immune regulatory factors. Retrospective analysis of clinical samples was conducted to assess the predictive and prognostic value of IRF1 in chemoimmunotherapy. Additionally, A549 cells with varying IRF1 expression levels were constructed to investigate its effects on NSCLC cells, while animal experiments were performed to study the role of IRF1 in vivo. Our findings revealed that the primary mutation of IRF1 is deep deletion and it exhibits a close association with immune regulatory factors. KRAS and TP53 are among the target genes of IRF1, with interferon and IL-2 being the predominantly affected pathways. Clinically, IRF1 levels significantly correlate with the efficacy of chemoimmunotherapy. Patients with high IRF1 levels exhibited a median progression-free survival (mPFS) of 9.5 months, whereas those with low IRF1 levels had a shorter mPFS of 5.8 months. IRF1 levels positively correlate with PD-L1 distribution and circulating IL-2 levels. IL-2 enhances the biological function of IRF1 and recapitulates its role in vivo in the knockdown group. Therefore, IRF1 may possess predictive and prognostic value for chemoimmunotherapy in NSCLC patients through the regulation of the IL-2 inflammatory pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:化学免疫疗法,这得益于化疗和免疫疗法的结合,已经成为癌症治疗中一种有前途的策略。然而,有效诱导强健的免疫应答仍然具有挑战性,因为患者之间的应答能力有限.内质网(ER)应激对于激活与免疫原性细胞死亡(ICD)相关的细胞内信号通路至关重要。针对ER的靶向药物可能会增强ER应激并改善ICD相关的免疫治疗.
    目的:提高化疗免疫疗法的免疫应答。
    方法:构建ER靶向纳米颗粒TSE-CEL/NP以增强免疫原性癌细胞死亡。流式细胞术,共聚焦显微镜,TEM和免疫荧光用于评估ER靶向作用和免疫原性肿瘤细胞对B16F10肿瘤细胞的体外死亡。构建单侧和双侧肿瘤模型以研究体内抗肿瘤和免疫疗法的功效。肺转移B16F10黑素瘤荷瘤小鼠用于评估抗转移功效。
    结果:TSE-CEL/NP可以在ER中特别积累,从而引起ER应激。高ER应力触发CRT的暴露,HMGB1和ATP的胞外释放。这些危险信号随后促进树突状细胞(DC)的募集和成熟,这反过来又增加了细胞毒性T淋巴细胞(CD8+T细胞)的增殖,最终改善了针对黑色素瘤的免疫疗法疗效.体内实验表明TSE-CEL/NP表现出优异的抗肿瘤功效并引发强烈的免疫应答。
    结论:我们的发现为黑色素瘤免疫治疗提供了新的ER靶向方法和实验基础。
    BACKGROUND: Chemoimmunotherapy, which benefits from the combination of chemotherapy and immunotherapy, has emerged as a promising strategy in cancer treatment. However, effectively inducing a robust immune response remains challenging due to the limited responsiveness across patients. Endoplasmic reticulum (ER) stress is essential for activating intracellular signaling pathways associated with immunogenic cell death (ICD), targeting drugs to ER might enhance ER stress and improve ICD-related immunotherapy.
    OBJECTIVE: To improve the immune response of Chemoimmunotherapy.
    METHODS: ER targeting nanoparticles TSE-CEL/NP were constructed to enhance immunogenic cancer cell death. Flow cytometry, confocal microscope, TEM and immunofluorescence were used to evaluate the ER targeting effect and immunogenic tumor cell death in vitro on B16F10 tumor cells. Unilateral and bilateral tumor models were constructed to investigate the efficacy of anti-tumor and immunotherapy in vivo. Lung metastasis B16F10 melanoma tumor-bearing mice were used to assess the anti-metastasis efficacy.
    RESULTS: TSE-CEL/NP could specially accumulate in ER, thereby induce ER stress. High ER stress trigger the exposure of CRT, the extracellular release of HMGB1 and ATP. These danger signals subsequently promote the recruitment and maturation of dendritic cells (DCs), which in turn increase the proliferation of cytotoxic T lymphocytes (CD8+ T cells), ultimately resulted in an improved immunotherapy efficacy against melanoma. Invivo experiments showed that TSE-CEL/NP exhibits excellent antitumor efficacy and triggers a strong immune response.
    CONCLUSIONS: Our findings demonstrated that celastrol ER targeting delivery could amplify immunogenic cell death in melanoma, which provide experimental basis for melanoma immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号