immune checkpoint blockade

免疫检查点阻断
  • 文章类型: Journal Article
    口腔鳞状细胞癌(OSCC)是全球范围内主要且危险的恶性肿瘤,大多数病例源于口腔潜在恶性疾病(OPMDs)。尽管如此,阻碍OPMD进展为OSCC的有效策略仍然难以捉摸。在这项研究中,我们通过4-硝基喹啉1-氧化物诱导建立小鼠口腔癌变模型,反映了从正常口腔粘膜到OPMD的顺序转变,最终发展到OSCC。通过在OPMD阶段进行干预,我们观察到,PD1阻断联合光动力疗法(PDT)可显著缓解口腔癌变进展.单细胞转录组测序揭示了主要从OPMD到OSCC阶段发生的微环境失调,促进以Treg比例增加为特征的肿瘤促进环境,增强S100A8表达式,并降低Fib_Igfbp5(一种特定的成纤维细胞亚型)的比例,在其他人中。值得注意的是,在OPMDs阶段干预PD1阻断和PDT阻碍了促进肿瘤微环境的形成,导致Treg比例下降,S100A8表达减少,增加了Fib_Igfbp5的比例。此外,与单药治疗相比,联合治疗引发的治疗相关免疫反应更为强劲.实质上,我们的发现为减少口腔癌变的进展提供了一种新的策略。
    Oral squamous cell carcinoma (OSCC) stands as a predominant and perilous malignant neoplasm globally, with the majority of cases originating from oral potential malignant disorders (OPMDs). Despite this, effective strategies to impede the progression of OPMDs to OSCC remain elusive. In this study, we established mouse models of oral carcinogenesis via 4-nitroquinoline 1-oxide induction, mirroring the sequential transformation from normal oral mucosa to OPMDs, culminating in OSCC development. By intervening during the OPMDs stage, we observed that combining PD1 blockade with photodynamic therapy (PDT) significantly mitigated oral carcinogenesis progression. Single-cell transcriptomic sequencing unveiled microenvironmental dysregulation occurring predominantly from OPMDs to OSCC stages, fostering a tumor-promoting milieu characterized by increased Treg proportion, heightened S100A8 expression, and decreased Fib_Igfbp5 (a specific fibroblast subtype) proportion, among others. Notably, intervening with PD1 blockade and PDT during the OPMDs stage hindered the formation of the tumor-promoting microenvironment, resulting in decreased Treg proportion, reduced S100A8 expression, and increased Fib_Igfbp5 proportion. Moreover, combination therapy elicited a more robust treatment-associated immune response compared with monotherapy. In essence, our findings present a novel strategy for curtailing the progression of oral carcinogenesis.
    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
    冷冻消融(Cryo)是一种针对肿瘤的微创治疗方法。低温可以激活人体的免疫反应,尽管它通常很弱。对Cryo在肝细胞癌(HCC)中诱导的免疫应答知之甚少。PD-1和CTLA-4单克隆抗体是用于肿瘤免疫治疗的免疫检查点抑制剂。这些抗体与Cryo的组合使用可以增强免疫效果。
    建立了Balb/c小鼠肝癌模型并用Cryo治疗,免疫检查点阻断(ICB),或冷冻+ICB(联合治疗)。确定了未治疗的肿瘤的生长趋势和小鼠的存活时间。Westernblot(WB)法检测细胞凋亡相关蛋白的表达。通过流式细胞术分析免疫细胞和免疫抑制细胞的百分比。通过免疫组织化学检查浸润T淋巴细胞的数量,通过定量实时聚合酶链反应(qRT-PCR)测定和酶联免疫吸附测定(ELISA)测定检测T细胞相关细胞因子的水平。
    冷冻+ICB抑制右侧未治疗肿瘤的生长,促进肿瘤细胞凋亡,延长了小鼠的存活时间。联合治疗后右侧肿瘤组织局部T细胞浸润增加,而免疫抑制细胞的数量显著减少。此外,联合治疗可诱导多种Th1型细胞因子的产生,但减少Th2型细胞因子的产生。
    冷冻可以激活CD8+和CD4+T细胞免疫应答。冷冻+ICB可以缓解免疫抑制肿瘤微环境,使Th1/Th2平衡向Th1优势转移,进一步增强了低温诱导的T细胞免疫应答,导致更强的抗肿瘤免疫应答。
    UNASSIGNED: Cryoablation (Cryo) is a minimally invasive treatment for tumors. Cryo can activate the body\'s immune response, although it is typically weak. The immune response induced by Cryo in hepatocellular carcinoma (HCC) is poorly understood. PD-1 and CTLA-4 monoclonal antibodies are immune checkpoint inhibitors used in immunotherapy for tumors. The combined use of these antibodies with Cryo may enhance the immune effect.
    UNASSIGNED: A Balb/c mouse model of HCC was established and treated with Cryo, immune checkpoint blockade (ICB), or Cryo + ICB (combination therapy). The growth trend of right untreated tumors and survival time of mice were determined. The expression of apoptosis-related proteins was detected by Western blot (WB) assay. The percentages of immune cells and immunosuppressive cells were analyzed by flow cytometry. The numbers of infiltrating T lymphocytes were checked by immunohistochemistry, and the levels of T-cell-associated cytokines were detected by Quantitative real-time Polymerase Chain Reaction (qRT-PCR) assays and Enzyme-Linked Immunosorbent Assays (ELISA) assays.
    UNASSIGNED: Cryo + ICB inhibited the growth of right untreated tumors, promoted tumor cell apoptosis, and prolonged the survival time of mice. Local T-cell infiltration in right tumor tissues increased after the combination therapy, while the number of immunosuppressive cells was significantly reduced. In addition, the combination therapy may induce the production of multiple Th1-type cytokines but reduce the production of Th2-type cytokines.
    UNASSIGNED: Cryo can activate CD8+ and CD4+ T-cell immune responses. Cryo + ICB can relieve the immunosuppressive tumor microenvironment and shift the Th1/Th2 balance toward Th1 dominance, further enhancing the Cryo-induced T-cell immune response and resulting in a stronger antitumor immune response.
    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
    光热疗法(PTT)是一种有前途的癌症治疗方法,因为它能够诱导肿瘤特异性T细胞反应并增强治疗效果。然而,不完全的PTT可留下残留的肿瘤,在临床情况下往往导致新的转移和降低患者生存率.这主要是由于ATP的释放,一种损伤相关的分子模式,通过CD39迅速转化为免疫抑制代谢物腺苷,在肿瘤微环境中普遍存在,从而促进肿瘤免疫逃避。这项研究提出了一种通过Fe掺杂的聚二氨基吡啶(Fe-PDAP)之间的静电吸附制备的光热纳米药物,吲哚菁绿(ICG),和CD39抑制剂聚氧钨酸钠(POM-1)。当暴露于近红外激光时,构建的Fe-PDAP@ICG@POM-1(FIP)可以诱导肿瘤PTT和免疫原性细胞死亡。重要的是,它可以通过双向免疫代谢调节抑制ATP-腺苷途径,导致ATP水平增加和腺苷合成减少,最终逆转免疫抑制微环境并增加免疫检查点阻断(aPD-1)治疗的敏感性。在aPD-1的帮助下,由FIP介导的双向免疫代谢调控策略可以有效地抑制/根除原发性和远端肿瘤,并唤起长期坚实的免疫记忆。这项研究提出了一种免疫代谢控制策略,为治疗不完全PTT后的残留肿瘤提供了挽救选择。
    Photothermal therapy (PTT) is a promising cancer treatment method due to its ability to induce tumor-specific T cell responses and enhance therapeutic outcomes. However, incomplete PTT can leave residual tumors that often lead to new metastases and decreased patient survival in clinical scenarios. This is primarily due to the release of ATP, a damage-associated molecular pattern that quickly transforms into the immunosuppressive metabolite adenosine by CD39, prevalent in the tumor microenvironment, thus promoting tumor immune evasion. This study presents a photothermal nanomedicine fabricated by electrostatic adsorption among the Fe-doped polydiaminopyridine (Fe-PDAP), indocyanine green (ICG), and CD39 inhibitor sodium polyoxotungstate (POM-1). The constructed Fe-PDAP@ICG@POM-1 (FIP) can induce tumor PTT and immunogenic cell death when exposed to a near-infrared laser. Significantly, it can inhibit the ATP-adenosine pathway by dual-directional immunometabolic regulation, resulting in increased ATP levels and decreased adenosine synthesis, which ultimately reverses the immunosuppressive microenvironment and increases the susceptibility of immune checkpoint blockade (aPD-1) therapy. With the aid of aPD-1, the dual-directional immunometabolic regulation strategy mediated by FIP can effectively suppress/eradicate primary and distant tumors and evoke long-term solid immunological memory. This study presents an immunometabolic control strategy to offer a salvage option for treating residual tumors following incomplete PTT.
    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
    基于免疫检查点抑制剂(ICI)的治疗在各种癌症类型中取得了令人印象深刻的成功。近十年来,几种ICI被空前批准为晚期肝细胞癌(HCC)的治疗方案。同时,正在进行许多临床试验,以利用更多的ICIs进入最初不可切除的HCC和术后HCC,以预期诱导足够的肿瘤降级以进一步切除或实施辅助治疗以实现无复发生存。分别。在这次审查中,我们的目的是总结一些语用的组织形态学,免疫组织化学,和分子病理参数有望表明新辅助/转化ICI相关治疗的反应,并预测辅助/治疗性ICI相关治疗对HCC的疗效。
    我们使用术语肝细胞癌搜索PubMed,免疫疗法,免疫检查点抑制剂,免疫检查点封锁,转换疗法,新辅助治疗,辅助治疗,生物标志物,病理评估,病理评估至2023年2月。
    尽管对相关HCC标本的病理评估尚无共识,令人鼓舞的是,一些研究集中在这个领域,而且,其他类型癌症的方法和参数也值得参考。对于接受免疫治疗的HCC标本的病理评估,一个合适的抽样方案,识别免疫疗法相关的病理反应,应强调病理反应率的量化。对于计划接受免疫治疗的HCC患者,肿瘤浸润淋巴细胞,肿瘤内三级淋巴结构,程序性细胞死亡配体1,Wnt/β-catenin,微卫星不稳定性和错配修复,肿瘤突变负荷和肿瘤新抗原,以及其他一些信号通路是ICI治疗反应的潜在预测生物标志物。
    免疫治疗时代的HCC管理出现了一个全新的病理学挑战,即提供与免疫治疗相关的诊断报告。尽管许多相关研究是临床前或不足的,它们可能在未来极大地改变HCC的免疫治疗策略。
    UNASSIGNED: Immune checkpoint inhibitor (ICI)-based therapy has achieved impressive success in various cancer types. Several ICIs have been unprecedentedly approved as the treatment regimens for advanced hepatocellular carcinoma (HCC) in recent decade. Meanwhile, numerous clinical trials are being performed to exploit more ICIs into initially unresectable HCC and postoperative HCC to expectantly induce adequate tumor downstaging for further resection or implement adjuvant treatment for relapse-free survival, respectively. In this review, we aim to summarize some pragmatic histomorphologic, immunohistochemical, and molecular pathologic parameters which promisingly indicate the response of neoadjuvant/conversion ICI-related therapy and predict the efficacy of adjuvant/therapeutic ICI-related therapy for HCC.
    UNASSIGNED: We searched PubMed using the terms hepatocellular carcinoma, immunotherapy, immune checkpoint inhibitor, immune checkpoint blockade, conversion therapy, neoadjuvant therapy, adjuvant therapy, biomarker, pathologic evaluation, pathologic assessment till February 2023.
    UNASSIGNED: Although there is no consensus regarding the pathologic evaluation of relevant HCC specimens, it is encouraging that a few of studies have concentrated on this field, and moreover, the methods and parameters noted on other cancer types are also worthy of reference. For the pathologic assessment of HCC specimens underwent immunotherapy, a suitable sampling scheme, identifying immunotherapy-related pathologic response, and quantification of pathologic response rate should be emphasized. For the patients of HCC who are scheduled to receive immunotherapy, tumor-infiltrating lymphocyte, intratumoral tertiary lymphoid structure, programmed cell death ligand 1, Wnt/β-catenin, microsatellite instability and mismatch repair, tumor mutational burden and tumor neoantigen, as well as some other signaling pathways are the potential predictive biomarkers of treatment response of ICI.
    UNASSIGNED: The management of HCC in the era of immunotherapy arises a brand-new pathological challenge that is to provide an immunotherapy-related diagnostic report. Albeit many related researches are preclinical or insufficient, they may tremendously alter the immunotherapy strategy of HCC in future.
    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
    尽管免疫检查点封锁(ICB)具有开创性的影响,非小细胞肺癌的反应率仍然适中,特别是在免疫排斥或免疫沙漠微环境中。Toll样受体7(TLR7)作为一个潜在的靶点桥接先天和适应性免疫,为联合治疗增加ICB疗效提供了一个有希望的途径。这里,我们探索了新型口服TLR7激动剂TQ-A3334的抗肿瘤活性及其在同基因鼠肺癌模型中通过组合策略增强抗程序性死亡配体1(PD-L1)治疗的潜力.口服TQ-A3334可显着减轻C57BL/6J小鼠的肿瘤负担,由I型干扰素(IFN)调节,并表现出低毒性。这种疗法在肿瘤组织中引起先天和适应性免疫细胞的激活。特别是通过I型IFN途径和随后的CXCL10表达增加CD8TIL的丰度。体外检查证实,IFN-α刺激的肿瘤细胞显示CXCL10分泌增加,有利于促进CD8T细胞的运输。此外,TQ-A3334与抗PD-L1联合治疗超过了肿瘤控制,与单一疗法相比,CD8+TIL频率进一步增加。这些结果表明,TQ-A3334可以动员先天免疫并促进T细胞募集到肿瘤微环境;TQ-A3334和抗PD-L1抗体的组合可以增强肿瘤对抗PD-L1治疗的敏感性。这证明了治疗免疫能力差的浸润性肺癌的巨大潜力。
    Despite the groundbreaking impact of immune checkpoint blockade (ICB), response rates in non-small cell lung cancer remain modest, particularly in immune-excluded or immune-desert microenvironments. Toll-like receptor 7 (TLR7) emerges as a latent target bridging innate and adaptive immunity, offering a promising avenue for combination therapies to augment ICB efficacy. Here, we explored the anti-tumor activity of the novel oral TLR7 agonist TQ-A3334 and its potential to enhance anti-programmed death ligand 1 (PD-L1) therapy through a combination strategy in a syngeneic murine lung cancer model. Oral administration of TQ-A3334 significantly alleviated tumor burden in C57BL/6J mice, modulated by type I interferon (IFN), and exhibited low toxicity. This therapy elicited activation of both innate and adaptive immune cells in tumor tissue, particularly increasing the abundance of CD8+ TILs through type I IFN pathway and subsequent CXCL10 expression. In vitro examinations validated that IFN-α-stimulated tumor cells exhibited increased secretion of CXCL10, conducive to the promoted trafficking of CD8+ T cells. Furthermore, combining TQ-A3334 with anti-PD-L1 treatment exceeded tumor control, with a further increase in CD8+ TIL frequency compared to monotherapy. These findings suggest that TQ-A3334 can mobilize innate immunity and promote T cell recruitment into the tumor microenvironment; a combination of TQ-A3334 and anti-PD-L1 antibodies can intensify the sensitivity of tumors to anti-PD-L1 therapy, which demonstrates significant potential for treating poorly immune-infiltrated lung cancer.
    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
    免疫检查点阻断导致了晚期胃癌治疗的突破。然而,胃癌的显著异质性,特别是肿瘤微环境的异质性,强调了抗肿瘤反应是多因素相互作用的反映。通过转录组学分析和动态血浆样本分析,我们确定了肿瘤微环境中的代谢“对峙”机制,如烟酰胺代谢的双重预后意义所示。具体来说,表达限速酶烟酰胺磷酸核糖基转移酶和烟酰胺N-甲基转移酶的巨噬细胞和成纤维细胞,分别,调节烟酰胺/1-甲基烟酰胺比例和CD8+T细胞功能。机械上,烟酰胺N-甲基转移酶被NOTCH途径转录因子RBP-J转录激活,并通过SIRT1/NICD轴进一步被包含烟酰胺磷酸核糖基转移酶的巨噬细胞衍生的细胞外囊泡抑制。通过自体注射细胞外囊泡操纵烟酰胺代谢恢复了胃癌中CD8+T细胞的细胞毒性和抗PD-1反应。
    Immune checkpoint blockade has led to breakthroughs in the treatment of advanced gastric cancer. However, the prominent heterogeneity in gastric cancer, notably the heterogeneity of the tumor microenvironment, highlights the idea that the antitumor response is a reflection of multifactorial interactions. Through transcriptomic analysis and dynamic plasma sample analysis, we identified a metabolic \"face-off\" mechanism within the tumor microenvironment, as shown by the dual prognostic significance of nicotinamide metabolism. Specifically, macrophages and fibroblasts expressing the rate-limiting enzymes nicotinamide phosphoribosyltransferase and nicotinamide N-methyltransferase, respectively, regulate the nicotinamide/1-methylnicotinamide ratio and CD8+ T cell function. Mechanistically, nicotinamide N-methyltransferase is transcriptionally activated by the NOTCH pathway transcription factor RBP-J and is further inhibited by macrophage-derived extracellular vesicles containing nicotinamide phosphoribosyltransferase via the SIRT1/NICD axis. Manipulating nicotinamide metabolism through autologous injection of extracellular vesicles restored CD8+ T cell cytotoxicity and the anti-PD-1 response in gastric cancer.
    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
    需要结合多模式方法的体外模型来剖析局部肿瘤免疫微环境(TIME)对免疫疗法的动态反应。在这里,患者来源的原发性肺癌类器官(pLCO)是通过分离肿瘤细胞簇产生的,包括浸润的免疫细胞.开发了一种功能相关的单细胞RNA测序(FascRNA-seq)平台,该平台允许在单类器官水平进行表型评估和scRNA-seq,以剖析单个pLCO的时间。对来自7名患者的171个个体pLCO的分析显示,pLCO保留了亲本肿瘤组织实质中的时间异质性,提供具有相同遗传背景但不同时间的模型。将单个pLCO的scRNA-seq数据与其对抗PD-1(αPD-1)免疫检查点阻断(ICB)的反应联系起来,可以确认CD8+T细胞在抗肿瘤免疫中的核心作用。用一组10个基因来鉴定潜在的肿瘤反应性T细胞,解开调节T细胞活性的因子,包括CD99基因。总之,本研究构建了一个联合表型和转录组FascRNA-seq平台,以剖析ICB处理下局部TIME的动态响应,提供了一种有前途的方法来评估新的免疫疗法和了解潜在的分子机制。
    In vitro models coupled with multimodal approaches are needed to dissect the dynamic response of local tumor immune microenvironment (TIME) to immunotherapy. Here the patient-derived primary lung cancer organoids (pLCOs) are generated by isolating tumor cell clusters, including the infiltrated immune cells. A function-associated single-cell RNA sequencing (FascRNA-seq) platform allowing both phenotypic evaluation and scRNA-seq at single-organoid level is developed to dissect the TIME of individual pLCOs. The analysis of 171 individual pLCOs derived from seven patients reveals that pLCOs retain the TIME heterogeneity in the parenchyma of parental tumor tissues, providing models with identical genetic background but various TIME. Linking the scRNA-seq data of individual pLCOs with their responses to anti-PD-1 (αPD-1) immune checkpoint blockade (ICB) allows to confirm the central role of CD8+ T cells in anti-tumor immunity, to identify potential tumor-reactive T cells with a set of 10 genes, and to unravel the factors regulating T cell activity, including CD99 gene. In summary, the study constructs a joint phenotypic and transcriptomic FascRNA-seq platform to dissect the dynamic response of local TIME under ICB treatment, providing a promising approach to evaluate novel immunotherapies and to understand the underlying molecular mechanisms.
    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:基于程序性细胞死亡-1/配体-1(PD-1/PD-L1)阻断的新辅助治疗是否可以使局部晚期癌基因突变的非小细胞肺癌(NSCLC)患者受益仍存在争议。这项回顾性研究旨在观察新辅助PD-1/PD-L1阻断联合化疗与化疗和相应的酪氨酸激酶抑制剂(TKIs)对可切除癌基因阳性NSCLC患者的疗效和安全性。
    方法:回顾性招募接受新辅助治疗的有癌基因改变的潜在可切除NSCLC患者,在同一时期内,我们对接受新辅助PD-(L)1阻断型新辅助治疗的癌基因阴性患者队列进行了回顾比较.主要目的是观察这些药物的疗效和无事件生存期(EFS)。安全概况,分子靶标,和免疫因素数据,包括PD-L1表达和肿瘤突变负担(TMB),也得到了。
    结果:共纳入46例患者。其中31个有癌基因改变,包括EGFR,KRAS,ERBB2,ROS1,MET,RET,ALK,和FGFR3改变。在癌基因阳性的患者中,18例患者接受新辅助PD-(L)1阻断免疫疗法加化疗(癌基因阳性IO组),13例患者接受新辅助化疗和/或相应的TKIs或TKIs单独治疗(癌基因阳性化疗/TKIs组),其他15例患者为癌基因阴性,接受新辅助PD-(L)1阻断加化疗(癌基因阴性IO组)。癌基因阳性IO组病理完全缓解(pCR)和主要病理缓解(MPR)率分别为22.2%(18个中的4个)和44.4%(18个中的8个),0%(P=0.120)和23.1%(13个中的3个)(P=0.276)在癌基因阳性化疗/TKIs组,癌基因阴性IO组的46.7%(15个中的7个)(P=0.163)和80.0%(15个中的12个)(P=0.072),分别。在最后的后续行动中,癌基因阳性IO组的中位EFS时间未达到,癌基因阳性化疗/TKIs组为29.5个月,癌基因阴性IO组为38.4个月。
    结论:与化疗/TKIs治疗相比,PD-(L)1阻断联合铂类化疗的新辅助治疗在部分可切除癌基因突变NSCLC患者中与较高的pCR/MPR率相关。而pCR/MPR率低于以PD-(L)1阻断为基础的治疗方案的癌基因阴性者。具体来说,在临床实践中,应考虑癌基因改变类型和免疫治疗反应的其他预测因子.
    BACKGROUND: Whether programmed cell death-1/ligand-1 (PD-1/PD-L1) blockade-based neoadjuvant treatment may benefit locally advanced oncogene-mutant non-small cell lung cancer (NSCLC) patients remains controversial. This retrospective study was designed to observe the efficacy and safety of neoadjuvant PD-1/PD-L1 blockade plus chemotherapy versus chemotherapy and corresponding tyrosine kinase inhibitors (TKIs) in patients with resectable oncogene-positive NSCLC.
    METHODS: Patients with potential resectable NSCLC harbouring oncogene alterations who had received neoadjuvant treatment were retrospectively recruited, and an oncogene-negative cohort of patients who received neoadjuvant PD-(L)1 blockade-based neoadjuvant treatment was reviewed for comparison during the same period. The primary aim was to observe the treatment efficacy and event-free survival (EFS) of these agents. Safety profile, molecular target, and immunologic factor data, including PD-L1 expression and tumour mutational burden (TMB), were also obtained.
    RESULTS: A total of 46 patients were recruited. Thirty-one of them harboured oncogene alterations, including EGFR, KRAS, ERBB2, ROS1, MET, RET, ALK, and FGFR3 alterations. Among the oncogene-positive patients, 18 patients received neoadjuvant PD-(L)1 blockade immunotherapy plus chemotherapy (oncogene-positive IO group), 13 patients were treated with neoadjuvant chemotherapy and/or corresponding TKIs or TKIs alone (oncogene-positive chemo/TKIs group), and the other 15 patients were oncogene negative and received neoadjuvant PD-(L)1 blockade plus chemotherapy (oncogene-negative IO group). The pathological complete response (pCR) and major pathological response (MPR) rates were 22.2% (4 of 18) and 44.4% (8 of 18) in the oncogene-positive IO group, 0% (P = 0.120) and 23.1% (3 of 13) (P = 0.276) in the oncogene-positive chemo/TKIs group, and 46.7% (7 of 15) (P = 0.163) and 80.0% (12 of 15) (P = 0.072) in the oncogene-negative IO group, respectively. By the last follow-up, the median EFS time had not reached in the oncogene-positive IO group, and was 29.5 months in the oncogene-positive chemo/TKIs group and 38.4 months in the oncogene-negative IO group.
    CONCLUSIONS: Compared with chemotherapy/TKIs treatment, neoadjuvant treatment with PD-(L)1 blockade plus platinum-based chemotherapy was associated with higher pCR/MPR rates in patients with partially resectable oncogene-mutant NSCLC, while the pCR/MPR rates were lower than their oncogene-negative counterparts treated with PD-(L)1 blockade-based treatment. Specifically, oncogene alteration types and other predictors of response to immunotherapy should be taken into account in clinical practice.
    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
    目前,声动力疗法(SDT)的治疗效果和免疫反应有限,强调迫切需要增强的策略,可以刺激强大和持久的抗肿瘤作用。微囊藻,臭名昭著的微藻,揭示了由于存在气体囊泡(GV)和藻蓝蛋白(PC)而介导SDT的可能性。在这里,一种无毒的elabens微囊藻(标记为Me)被开发为SDT的新型药剂,因为它在红光照射下产生O2,而GV和PC充当空化核和声敏剂,分别。此外,超声波照射后释放藻类碎片,启动Toll样受体途径以启动一系列免疫反应。这种声免疫策略主要通过促进树突状细胞成熟和细胞毒性T细胞活化来抑制CT26结肠肿瘤生长。与免疫检查点阻断结合后,治疗结果进一步放大,伴随着令人满意的切除和免疫记忆效果;在“冷”4T1三阴性乳腺肿瘤中证明了类似的效力。此外,Me表现出良好的生物安全性,没有明显的急性或慢性毒性。简而言之,这项研究通过引入基于微囊藻的声免疫疗法将废物转化为财富,对癌症取得了令人鼓舞的治疗效果,预计将被翻译成诊所。本文受版权保护。保留所有权利。
    Currently, sonodynamic therapy (SDT) has limited therapeutic outcomes and immune responses, highlighting the urgent need for enhanced strategies that can stimulate robust and long-lasting antitumor effects. Microcystis, a notorious microalga, reveals the possibility of mediating SDT owing to the presence of gas vesicles (GVs) and phycocyanin (PC). Herein, a nontoxic strain of Microcystis elabens (labeled Me) is developed as a novel agent for SDT because it generates O2 under red light (RL) illumination, while GVs and PC act as cavitation nuclei and sonosensitizers, respectively. Moreover, algal debris is released after ultrasound (US) irradiation, which primes the Toll-like receptor pathway to initiate a cascade of immune responses. This sono-immune strategy inhibits CT26 colon tumor growth largely by promoting dendritic cell (DC) maturation and cytotoxic T-cell activation. After combination with the immune checkpoint blockade (ICB), the therapeutic outcome is further amplified, accompanied by satisfactory abscopal and immune memory effects; the similar potency is proven in the \"cold\" 4T1 triple-negative breast tumor. In addition, Me exhibits good biosafety without significant acute or chronic toxicity. Briefly, this study turns waste into wealth by introducing sono-immunotherapy based on Microcystis that achieved encouraging therapeutic effects on cancer, which is expected to be translated into the clinic.
    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
    基于时空控释和肿瘤成像整合的智能药物平台有望克服传统治疗模式的低效率和不确定性。在这项研究中,开发了一种由热敏水凝胶(聚乙烯醇-羧酸水凝胶(PCF))和多功能纳米颗粒(Fe3O4@Au/Mn(Zn)-4-羧基苯基卟啉/聚多巴胺(FAMxP))组成的复合材料,以在磁共振成像(MRI)和荧光成像(FI)的指导下结合肿瘤免疫原性细胞死亡(ICD)/免疫阻滞(ICB)治疗。它不仅可以通过肿瘤细胞的叶酸受体进一步识别靶细胞,而且在暴露于近红外光后也会产生热溶解,从而在原位缓慢释放FAMxP,从而延长治疗时间,避免肿瘤复发。当FAMxP进入肿瘤细胞时,它以pH依赖性方式释放FAMx。化学动力学,光热和光动力疗法可在癌细胞中引起显著的ICD。因此,通过注射抗程序性细胞死亡配体1可以进一步增强ICB,从而提高肿瘤治疗的有效性。开发的PCF-FAMxP复合水凝胶可以代表具有用于肿瘤的协作MRI/FI引导的靶向治疗途径的简单组合物的更新的药物设计方法。
    Smart drug platforms based on spatiotemporally controlled release and integration of tumor imaging are expected to overcome the inefficiency and uncertainty of traditional theranostic modes. In this study, a composite consisting of a thermosensitive hydrogel (polyvinyl alcohol-carboxylic acid hydrogel (PCF)) and a multifunctional nanoparticle (Fe3O4@Au/Mn(Zn)-4-carboxyphenyl porphyrin/polydopamine (FAMxP)) is developed to combine tumor immunogenic cell death (ICD)/immune checkpoint blockade (ICB) therapy under the guidance of magnetic resonance imaging (MRI) and fluorescence imaging (FI). It can not only further recognize the target cells through the folate receptor of tumor cells, but also produce thermal dissolution after exposure to near-infrared light to slowly release FAMxP in situ, thereby prolonging the treatment time and avoiding tumor recurrence. As FAMxP entered the tumor cells, it released FAMx in a pH-dependent manner. Chemodynamic, photothermal and photodynamic therapy can cause significant ICD in cancer cells. ICB can thus be further enhanced by injecting anti-programmed cell death ligand 1, improving the effectiveness of tumor treatment. The developed PCF-FAMxP composite hydrogel may represent an updated drug design approach with simple compositions for cooperative MRI/FI-guided targeted therapeutic pathways for tumors.
    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