Immune checkpoint blockade

免疫检查点阻断
  • 文章类型: Journal Article
    治疗诱导的免疫原性细胞死亡(ICD)和免疫检查点阻断的组合可以提供相互增强的策略来逆转肿瘤的不良免疫原性和免疫逃逸行为。在这项工作中,嵌合肽工程免疫刺激剂(ER-PPB)被制造用于针对转移性肿瘤的内质网(ER)靶向光动力免疫疗法。其中,两亲性嵌合肽(ER-PP)由ER靶向肽FFKDEL组成,亲水PEG8接头和光敏剂原卟啉IX(PpIX),可以与PD-1/PD-L1阻断剂(BMS-1)组装以制备ER-PPB。被动靶向肿瘤组织后,ER-PPB将选择性地积累在ER中。接下来,ER-PPB的局部PDT会产生大量的ROS破坏原发肿瘤细胞,同时增加ER应力以启动强大的ICD级联。此外,BMS-1的伴随递送可以通过PD-1/PD-L1阻断阻止肿瘤细胞的免疫逃逸,从而协同激活免疫系统来对抗转移性肿瘤。体外和体内结果表明ER-PPB具有强大的免疫激活和转移性肿瘤抑制特征,这可能为时空控制的转移性肿瘤治疗提供有希望的策略。
    The combination of therapy-induced immunogenic cell death (ICD) and immune checkpoint blockade can provide a mutually reinforced strategy to reverse the poor immunogenicity and immune escape behavior of tumors. In this work, a chimeric peptide-engineered immunostimulant (ER-PPB) is fabricated for endoplasmic reticulum (ER)-targeted photodynamic immunotherapy against metastatic tumors. Among which, the amphiphilic chimeric peptide (ER-PP) is composed of ER-targeting peptide FFKDEL, hydrophilic PEG8 linker and photosensitizer protoporphyrin IX (PpIX), which could be assembled with a PD-1/PD-L1 blocker (BMS-1) to prepare ER-PPB. After passively targeting at tumor tissues, ER-PPB will selectively accumulate in the ER. Next, the localized PDT of ER-PPB will produce a lot of ROS to destroy the primary tumor cells, while increasing the ER stress to initiate a robust ICD cascade. Moreover, the concomitant delivery of BMS-1 can impede the immune escape of tumor cells through PD-1/PD-L1 blockade, thus synergistically activating the immune system to combat metastatic tumors. In vitro and in vivo results demonstrate the robust immune activation and metastatic tumor inhibition characteristics of ER-PPB, which may offer a promising strategy for spatiotemporally controlled metastatic tumor therapy.
    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
    由于免疫原性不足和高度免疫抑制的肿瘤微环境(TME),三阴性乳腺癌(TNBC)对免疫疗法的反应较差。在这里,一种智能钙/钴基纳米调节剂(Ca,Co)CO3-LND-TCPP@F127-TA(缩写为CCLT@FT)被开发用作声动力学铁凋亡诱导剂和代谢免疫佐剂,以增强抗肿瘤免疫疗法。更多细节,由于CCLT@FT中Co2/Co3氧化还原对的存在,可以同时产生活性氧(ROS)和消耗谷胱甘肽(GSH)。同时,线粒体Ca2过载和四(4-羧基苯基)卟啉(TCPP)介导的声动力疗法(SDT)进一步放大了氧化应激并促进了肿瘤细胞的铁凋亡。更令人印象深刻的是,CCLT@FT可以通过掺杂钴和负载lonidamine来调节乳酸代谢(LND,MCT4的抑制剂),从而逆转高乳酸免疫抑制TME。此外,与免疫检查点阻断(ICB)治疗的组合被发现可以实现优异的抗肿瘤免疫力,反过来通过下调SLC7A11蛋白促进肿瘤细胞的铁凋亡,最终创造了一个“循环”疗法。总的来说,这项工作证明了一种基于CCLT@FT诱导铁凋亡/乳酸代谢调节和ICB治疗之间的闭环增强治疗途径的增强抗肿瘤免疫治疗的新策略,为TNBC的有效免疫治疗提供了替代和重要的参考。
    Triple-negative breast cancer (TNBC) responds poorly to immunotherapy due to insufficient immunogenicity and highly immunosuppressive tumor microenvironment (TME). Herein, an intelligent calcium/cobalt-based nanomodulator (Ca,Co)CO3-LND-TCPP@F127-TA (abbreviated as CCLT@FT) is developed to act as a sonodynamic-ferroptosis inducer and metabolic immunoadjuvant to enhance anti-tumor immunotherapy. More details, simultaneous reactive oxygen species (ROS) generation and glutathione (GSH) depletion can be achieved due to the existence of Co2+/Co3+ redox couple in CCLT@FT. Meanwhile, mitochondrial Ca2+ overload and tetrakis(4-carboxyphenyl) porphyrin (TCPP)-mediated sonodynamic therapy (SDT) further amplify the oxidative stress and promote ferroptosis in tumor cells. More impressively, CCLT@FT can modulate lactate metabolism by doping with cobalt and loading with lonidamine (LND, an inhibitor of MCT4), thereby reversing the high-lactate immunosuppressive TME. Furthermore, the combination with immune checkpoint blockade (ICB) therapy is found to achieve superior anti-tumor immunity, which in turn promotes ferroptosis of tumor cells by downregulating SLC7A11 protein, ultimately creating a \"cycle\" therapy. Overall, this work demonstrates a novel strategy for enhancing anti-tumor immunotherapy based on a closed-loop enhancement therapeutic route between CCLT@FT inducing ferroptosis/lactate metabolism modulation and ICB therapy, providing an alternative and important reference for effective immunotherapy of TNBC.
    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
    IFNγ,一种多效性细胞因子,不仅由活化的淋巴细胞产生,而且还响应于癌症免疫疗法,具有抗肿瘤和促进肿瘤的功能。在卵巢癌(OC)细胞中,IFNγ的肿瘤促进功能是由IFNγ诱导的Bcl3,PD-L1和IL-8/CXCL8的表达介导的,长期以来,这些基因作为原癌基因具有关键的细胞功能,免疫检查点配体和化学引诱物,分别。然而,大量证据表明,这三个基因具有远远超出其最初确定的功能的促肿瘤作用。这些肿瘤促进机制包括增加癌细胞增殖,入侵,血管生成,转移,化疗抗性和免疫逃逸。最近的研究表明,IFNγ诱导的Bcl3、PD-L1和IL-8的表达受相同的JAK1/STAT1信号通路的调控:IFNγ诱导Bcl3的表达,进而促进OC细胞中PD-L1和IL-8的表达,导致它们的扩散和迁移增加。在这次审查中,我们总结了IFNγ如何影响肿瘤微环境并促进肿瘤进展的最新发现,特别关注卵巢癌和Bcl3,PD-L1和IL-8/CXCL8信号传导。我们还讨论了针对Bcl3,PD-L1和IL-8的临床试验中的有希望的新型组合策略,以提高癌症免疫疗法的有效性。
    IFNγ, a pleiotropic cytokine produced not only by activated lymphocytes but also in response to cancer immunotherapies, has both antitumor and tumor-promoting functions. In ovarian cancer (OC) cells, the tumor-promoting functions of IFNγ are mediated by IFNγ-induced expression of Bcl3, PD-L1 and IL-8/CXCL8, which have long been known to have critical cellular functions as a proto-oncogene, an immune checkpoint ligand and a chemoattractant, respectively. However, overwhelming evidence has demonstrated that these three genes have tumor-promoting roles far beyond their originally identified functions. These tumor-promoting mechanisms include increased cancer cell proliferation, invasion, angiogenesis, metastasis, resistance to chemotherapy and immune escape. Recent studies have shown that IFNγ-induced Bcl3, PD-L1 and IL-8 expression is regulated by the same JAK1/STAT1 signaling pathway: IFNγ induces the expression of Bcl3, which then promotes the expression of PD-L1 and IL-8 in OC cells, resulting in their increased proliferation and migration. In this review, we summarize the recent findings on how IFNγ affects the tumor microenvironment and promotes tumor progression, with a special focus on ovarian cancer and on Bcl3, PD-L1 and IL-8/CXCL8 signaling. We also discuss promising novel combinatorial strategies in clinical trials targeting Bcl3, PD-L1 and IL-8 to increase the effectiveness of cancer immunotherapies.
    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:立体定向放射治疗(SBRT)对于颅外转移性疾病的治疗是安全有效的,但其与免疫检查点抑制剂(ICI)联合使用时的安全性尚未得到全面报道。在这里,我们报告了与SBRT和ICI联合相关的不良事件(AE),使用三个研究多位点SBRT联合ICI的前瞻性收集的患者数据。
    方法:患者纳入了3项ICI前瞻性试验(pembrolizumab;nivolumab/urelumab或nivolumab/cabiralizumab;nivolumab/ipilimumab),SBRT至1-4个位点。使用CTCAEv4.0前瞻性记录AE。使用具有90天标志的Kaplan-Meier方法分析存活。使用Fine-Gray回归评估患者特征与AE累积发生率的关联。
    结果:纳入213例患者,中位随访时间为10个月。在随访期间,50%和27%的患者经历至少一个≥2级或≥3级AE,分别。6个月≥2级和≥3级不良事件的累积发生率分别为47%和23%,分别。在90天剂量限制性毒性窗口之外发生了与治疗相关的“可能”等级5级AE。有或没有≥3级AEs的患者的标志生存分析显示,无进展生存或总生存没有显着差异。双药ICI与≥3级AE显著相关。
    结论:本分析的特点是迄今为止最大的前瞻性评估队列患者接受消融性SBRT和ICI联合治疗,并为未来的试验设计提供了背景。我们得出的结论是,在优先考虑正常组织约束的情况下交付SBRT时,可以安全地共同施用多位点SBRT和ICI。
    BACKGROUND: Stereotactic body radiotherapy (SBRT) is safe and effective for treatment of extracranial metastatic disease, but its safety when combined with immune checkpoint inhibitors (ICI) has not yet been comprehensively reported. Here we report adverse events (AEs) associated with combined SBRT and ICI using prospectively-collected data on patients in three trials investigating multi-site SBRT combined with ICI.
    METHODS: Patients were included from three prospective trials of ICI (pembrolizumab; nivolumab/urelumab or nivolumab/cabiralizumab; nivolumab/ipilimumab) with SBRT to 1-4 sites. AEs were recorded prospectively using the CTCAE v4.0. Survival was analyzed using Kaplan-Meier method with a 90-day landmark. Association of patient characteristics with cumulative incidence of AEs was assessed using Fine-Gray regression.
    RESULTS: 213 patients were included, with a median follow-up of 10 months. Over the follow-up period, 50 % and 27 % of patients experienced at least one grade ≥ 2 or grade ≥ 3 AE, respectively. Cumulative incidences of grade ≥ 2 and grade ≥ 3 AEs at 6 months were 47 % and 23 %, respectively. Three grade 5 AEs rated \"possibly\" related to treatment occurred outside the 90-day dose-limiting toxicity window. Landmarked survival analysis of patients with or without grade ≥ 3 AEs showed no significant difference in progression-free or overall survival. Dual-agent ICI was significantly associated with grade ≥ 3 AE.
    CONCLUSIONS: This analysis features the largest prospectively evaluated cohort of patients treated with combination ablative SBRT and ICI to date and provides context for future trial design. We conclude that multi-site SBRT and ICI can be safely co-administered when SBRT is delivered with prioritization of normal tissue constraints.
    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(ICB)疗法已在癌症类型中实现了广泛的适用性和持久的临床反应。然而,总体缓解率仍然不理想,因为一些患者没有缓解或出现耐药性.由于抗原呈递不足,CD8细胞毒性T细胞(CTL)在肿瘤微环境中的低浸润与对ICB的先天抗性密切相关。细胞表面上主要组织相容性复合物I类(MHC-I)表达的持续时间和空间分布对于内源性肿瘤抗原的有效呈递以及随后的CTL识别和清除至关重要。肿瘤细胞通过多种机制减少MHC-I的表面表达,从而削弱抗原呈递途径并逃避免疫和/或对ICB治疗产生抗性。随着越来越多的研究集中在膜MHC-I在肿瘤细胞中的运输和降解,这可能会影响肿瘤免疫治疗的有效性。有必要总结通过溶酶体调节膜MHC-I易位进入细胞质和降解的机制。我们回顾了对内体-溶酶体MHC-I转运的理解的最新进展,并强调了肿瘤细胞逃避CTL检测和清除的手段。我们还总结了针对这些途径的新治疗策略,以增强经典的ICB治疗,并为优化癌症免疫治疗提供了新的途径。
    Immune checkpoint blockade (ICB) therapy has achieved broad applicability and durable clinical responses across cancer types. However, the overall response rate remains suboptimal because some patients do not respond or develop drug resistance. The low infiltration of CD8+ cytotoxic T cells (CTLs) in the tumor microenvironment due to insufficient antigen presentation is closely related to the innate resistance to ICB. The duration and spatial distribution of major histocompatibility complex class I (MHC-I) expression on the cell surface is critical for the efficient presentation of endogenous tumor antigens and subsequent recognition and clearance by CTLs. Tumor cells reduce the surface expression of MHC-I via multiple mechanisms to impair antigen presentation pathways and evade immunity and/or develop resistance to ICB therapy. As an increasing number of studies have focused on membrane MHC-I trafficking and degradation in tumor cells, which may impact the effectiveness of tumor immunotherapy. It is necessary to summarize the mechanism regulating membrane MHC-I translocation into the cytoplasm and degradation via the lysosome. We reviewed recent advances in the understanding of endosomal-lysosomal MHC-I transport and highlighted the means exploited by tumor cells to evade detection and clearance by CTLs. We also summarized new therapeutic strategies targeting these pathways to enhance classical ICB treatment and provide new avenues for optimizing cancer 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:免疫检查点阻断(ICB)的治疗潜力遍及各种癌症;然而,其治疗肝细胞癌(HCC)的有效性经常受到固有和已发展的耐药性的削弱。
    目的:本研究探讨了将安洛替尼(一种广谱酪氨酸激酶抑制剂)与程序性死亡-1(PD-1)阻断相结合的有效性,并为更有效的治疗HCC策略提供了机制见解。
    方法:使用患者来源的器官型组织球体和原位HCC小鼠模型,我们评估了安洛替尼联合PD-1阻断的有效性.对肿瘤免疫微环境和潜在机制的影响使用飞行时间质量细胞计数评估,RNA测序,和跨细胞系的蛋白质组学,小鼠模型,和HCC患者样本。
    结果:在临床前模型中,安洛替尼与抗PD-1抗体的组合增强了针对HCC的免疫应答。安洛替尼通过VEGFR2/AKT/HIF-1α信号轴显著抑制转铁蛋白受体(TFRC)的表达。CD8+T细胞浸润到肿瘤微环境中与TFRC的低表达相关。安洛替尼还增加了趋化因子CXCL14的水平,这对于吸引CD8+T细胞至关重要。CXCL14成为TFRC的下游效应物,TFRC沉默后表现出表达升高。重要的是,低TFRC表达也与更好的预后相关,增强对联合治疗的敏感性,和肝癌患者抗PD-1治疗的良好反应。
    结论:我们的发现强调了安洛替尼通过靶向TFRC和增强CXCL14介导的CD8+T细胞浸润来增强抗PD-1免疫治疗在HCC中的疗效的潜力。这项研究有助于开发新的肝癌治疗策略,强调精准医学在肿瘤学中的作用。
    结论:在HCC临床前模型中证明了安洛替尼和抗PD-1免疫疗法的协同作用。安洛替尼通过VEGFR2/AKT/HIF-1α途径抑制TFRC表达。CXCL14通过TFRC抑制上调促进CD8+T细胞募集。TFRC成为评估晚期HCC患者预后和预测基于抗PD-1疗法反应的潜在生物标志物。
    BACKGROUND: The therapeutic potential of immune checkpoint blockade (ICB) extends across various cancers; however, its effectiveness in treating hepatocellular carcinoma (HCC) is frequently curtailed by both inherent and developed resistance.
    OBJECTIVE: This research explored the effectiveness of integrating anlotinib (a broad-spectrum tyrosine kinase inhibitor) with programmed death-1 (PD-1) blockade and offers mechanistic insights into more effective strategies for treating HCC.
    METHODS: Using patient-derived organotypic tissue spheroids and orthotopic HCC mouse models, we assessed the effectiveness of anlotinib combined with PD-1 blockade. The impact on the tumour immune microenvironment and underlying mechanisms were assessed using time-of-flight mass cytometry, RNA sequencing, and proteomics across cell lines, mouse models, and HCC patient samples.
    RESULTS: The combination of anlotinib with an anti-PD-1 antibody enhanced the immune response against HCC in preclinical models. Anlotinib remarkably suppressed the expression of transferrin receptor (TFRC) via the VEGFR2/AKT/HIF-1α signaling axis. CD8+ T-cell infiltration into the tumour microenvironment correlated with low expression of TFRC. Anlotinib additionally increased the levels of the chemokine CXCL14, crucial for attracting CD8+ T cells. CXCL14 emerged as a downstream effector of TFRC, exhibiting elevated expression following the silencing of TFRC. Importantly, low TFRC expression was also associated with a better prognosis, enhanced sensitivity to combination therapy, and a favourable response to anti-PD-1 therapy in patients with HCC.
    CONCLUSIONS: Our findings highlight anlotinib\'s potential to augment the efficacy of anti-PD-1 immunotherapy in HCC by targeting TFRC and enhancing CXCL14-mediated CD8+ T-cell infiltration. This study contributes to developing novel therapeutic strategies for HCC, emphasizing the role of precision medicine in oncology.
    CONCLUSIONS: Synergistic effects of anlotinib and anti-PD-1 immunotherapy demonstrated in HCC preclinical models. Anlotinib inhibits TFRC expression via the VEGFR2/AKT/HIF-1α pathway. CXCL14 upregulation via TFRC suppression boosts CD8+ T-cell recruitment. TFRC emerges as a potential biomarker for evaluating prognosis and predicting response to anti-PD-1-based therapies in advanced HCC 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
    免疫检查点抑制剂(ICIs)和酪氨酸激酶抑制剂(TKIs)的组合可与显著的毒性相关。我们在实体器官恶性肿瘤的临床试验中,对ICIs与TKIs(ICITKI)联合治疗的毒性进行了系统评价和荟萃分析。我们的主要终点调查了3-5级(G3-5)治疗相关毒性的发生率,我们的次要终点包括治疗类型的毒性发生率,疾病类型和磨合策略研究。总共确定了9750份摘要,其中包括72项符合条件的研究.最常见的疾病类型是非小细胞肺癌(n=8,11.1%),肾细胞癌(n=10,13.8%)和肝胆类癌(n=10,13.8%)。G3-5毒性的总发生率为56%(95%CI=50%-61%)。在这项分析中,最常见的TKIs与ICIs结合是多目标TKIs(n=52,72%),VEGF特异性(n=9,12.5%),或靶向癌基因的TKIs(EGFR,ALK,BRAF,MEK)(n=11,15.3%)。以癌基因为目标的TKIs与皮疹和免疫相关不良事件(irAEs)的发生率较高以及高血压的发生率较低相关。在使用TKI“磨合”减轻毒性的研究中,G3-5毒性的汇总估计值为71%(95%CI57-81%).几乎一半的研究(48%)忽略了G3-5irAE的发生率。我们的工作表明,接受ICI-TKI组合的大多数患者将经历高度毒性(G3-G5),并且毒性可能对TKI伴侣具有特异性(癌基因靶向TKIs:皮疹,irAE;VEGF/多靶向:高血压)。这些数据并不表明TKI“磨合”与G3-5毒性的较低发生率相关。irAE的报告不一致,支持需要协调不良事件报告以包括发病,持续时间和治疗。
    https://www.crd.约克。AC.英国/普华永道/,标识符CRD42022367416。
    The combination of immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) can be associated with significant toxicity. We performed a systematic review and meta-analysis of the toxicity of combination treatment of ICIs with TKIs (ICI + TKI) in clinical trials with solid organ malignancies. Our primary endpoint explored the incidence of grade 3 - 5 (G3-5) treatment-related toxicity and our secondary endpoints included the incidence of toxicity by treatment type, disease type and studies with run-in strategies. A total of 9750 abstracts were identified, of which 72 eligible studies were included. The most common disease types were non-small cell lung cancer (n=8, 11.1%), renal cell carcinoma (n=10, 13.8%) and hepatobiliary cancers (n=10, 13.8%). The overall incidence of G3-5 toxicity was 56% (95% CI = 50% - 61%). The most common TKIs combined with ICIs in this analysis were multi-targeted TKIs (n = 52, 72%), VEGF specific (n = 9, 12.5%), or oncogene-targeting TKIs (EGFR, ALK, BRAF, MEK) (n =11, 15.3%). Oncogene-targeted TKIs were associated a higher incidence of rashes and immune related adverse events (irAEs) and lower incidence of hypertension. In studies which used a TKI \'run-in\' to mitigate toxicity, the pooled estimate of G3-5 toxicity was 71% (95% CI 57-81%). Almost half of studies (48%) omitted the incidence of G3-5 irAEs. Our work suggests that the majority of patients who receive ICI-TKI combinations will experience high grade toxicity (G3-G5) and that toxicity may be specific to TKI partner (Oncogene targeted TKIs: Rash, irAEs; VEGF/Multitargeted: Hypertension). These data did not suggest that a TKI \'run-in\' was associated with a lower incidence of G3-5 toxicity. Reporting of irAEs was inconsistent supporting the need for harmonisation of adverse event reporting to include onset, duration and treatment.
    UNASSIGNED: https://www.crd.york.ac.uk/prospero/, identifier CRD42022367416.
    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
    由于抗原递送和呈递效率低下,目前癌症疫苗研究的临床转化受到抗肿瘤免疫反应有限的阻碍。次优的DC和T细胞激活。基于生物材料的纳米疫苗提供靶向抗原递送,保护免受体内降解,和延长肿瘤治疗效果。本研究引入了脂质包被的脱氧胆酸-存活素纳米组装体(DA-L-DSA)。幸存者,在几种癌细胞中过表达,并参与癌细胞生长和免疫逃避,被选为肿瘤相关抗原。survivin的主要组织相容性复合物I类结合表位被工程化到纳米组装体中。R848,TLR7/8激动剂,和SD-208,TGF-β受体1激酶抑制剂,共包封到纳米组装体中作为有效的佐剂以促进DC成熟和增强抗原呈递。DA-L-DSA有效刺激树突状细胞的成熟,迁移到淋巴结,并增强T细胞活化和Th1应答。在鼠黑素瘤模型中观察到细胞毒性T淋巴细胞大量流入原发性肿瘤,并在自发性乳腺癌转移模型中证明了抗转移作用。此外,DA-L-DSA在与免疫检查点抑制剂的联合治疗中表现出显著的协同作用,从而减轻免疫抑制性肿瘤微环境。一起来看,这些研究结果表明,DA-L-DSA是一种有前景的免疫治疗平台,可适用于多种顽固性癌症.
    Clinical translation of current cancer vaccine research has been hampered by limited antitumor immune responses due to inefficient antigen delivery and presentation, suboptimal DC and T cell activation. Biomaterial-based nanovaccine offers targeted antigen delivery, protection from degradation in vivo, and prolonged tumor therapeutic efficacy. This study introduces a lipid-coated deoxycholic acid-survivin nanoassembly (DA-L-DSA). Survivin, overexpressed in several cancer cells and involved in cancer cell growth and immune evasion, is selected as a tumor-associated antigen. An major histocompatibility complex class I binding epitope of survivin is engineered into the nanoassembly. R848, TLR 7/8 agonist, and SD-208, TGF-beta receptor1 kinase inhibitor, are coencapsulated into the nanoassembly as potent adjuvants to boost DC maturation and enhance antigen presentation. The DA-L-DSA effectively stimulates the maturation of dendritic cells, migrates into lymph nodes, and enhances T-cell activation and Th1 response. A substantial influx of cytotoxic T lymphocytes into primary tumors is observed in a murine melanoma model and demonstrates anti-metastatic effects in a spontaneous breast cancer metastasis model. Furthermore, DA-L-DSA exhibits a remarkable synergistic effect in the combination therapy with immune checkpoint inhibitors alleviating immunosuppressive tumor microenvironment. Taken together, these findings suggest DA-L-DSA as a promising immuno-therapeutic platform that could be applicable to diverse intractable cancers.
    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
    黑色素瘤仍然是美国最常见的癌症之一,然而,在可切除疾病的治疗方面取得了实质性进展。免疫检查点阻断(ICB)的辅助治疗和BRAF/MEK抑制剂(BRAF/MEKi)的靶向治疗现已成为可切除IIIB-IV期黑色素瘤的标准治疗。在这篇文章中,作者讨论了与可切除的黑色素瘤包括ICB治疗相关的最新科学发展,BRAF/MEKi靶向治疗,溶瘤病毒,肿瘤浸润淋巴细胞治疗,和癌症疫苗。
    Melanoma remains one of the most common cancers diagnosed in the United States, yet there have been substantial advancements in the treatment of resectable disease. Adjuvant therapy with immune checkpoint blockade (ICB) and targeted therapy with BRAF/MEK inhibitors (BRAF/MEKi) have now become standard of care for resectable stage IIIB-IV melanoma. In this article, the authors discuss recent scientific developments pertinent to the treatment of resectable melanoma including ICB, targeted therapy with BRAF/MEKi, oncolytic viruses, tumor-infiltrating lymphocyte therapy, and cancer vaccines.
    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
    确定将受益于5-氟尿嘧啶(5-FU)为基础的辅助化疗的II期和III期结肠癌患者对于推进个性化癌症治疗至关重要。我们采用半监督机器学习方法来分析具有933个II期和III期结肠癌样本的大型数据集。我们的分析利用基因调控网络来发现18个基因的预后特征,并探索可能预测化疗益处的10个基因特征。10个基因标记显示出强大的预后能力,并显示出预测化疗益处的潜力。我们在NanoStringnCounter平台上建立了强大的临床检测方法,在回顾性福尔马林固定石蜡包埋(FFPE)队列中验证,这是迈向临床应用的重要一步。我们的研究为改善结肠癌的辅助化疗和潜在的免疫治疗决策奠定了基础。需要未来的前瞻性研究来验证和建立10基因签名在临床环境中的临床实用性。
    Identifying patients with stage II and III colon cancer who will benefit from 5-fluorouracil (5-FU)-based adjuvant chemotherapy is crucial for the advancement of personalized cancer therapy. We employ a semi-supervised machine learning approach to analyze a large dataset with 933 stage II and III colon cancer samples. Our analysis leverages gene regulatory networks to discover an 18-gene prognostic signature and to explore a 10-gene signature that potentially predicts chemotherapy benefits. The 10-gene signature demonstrates strong prognostic power and shows promising potential to predict chemotherapy benefits. We establish a robust clinical assay on the NanoString nCounter platform, validated in a retrospective formalin-fixed paraffin-embedded (FFPE) cohort, which represents an important step toward clinical application. Our study lays the groundwork for improving adjuvant chemotherapy and potentially expanding into immunotherapy decision-making in colon cancer. Future prospective studies are needed to validate and establish the clinical utility of the 10-gene signature in clinical settings.
    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