Immunoradiotherapy

  • 文章类型: Journal Article
    背景:TLR7是抗病毒免疫的关键角色。TLR7信号传导激活抗原呈递细胞,包括DC和巨噬细胞。这种激活导致包括T细胞和B细胞的适应性免疫。因此,TLR7是免疫系统的重要分子。基于这些观察,TLR7激动剂被认为是一种使免疫系统对抗癌症的疗法。放射治疗(RT)是标准的癌症治疗方法之一,据报道可调节肿瘤免疫反应。在这项研究中,我们的目的是研究TLR7激动剂组合的抗肿瘤活性,DSP-0509,具有RT和底层机制。
    结果:我们表明,在CT26,LM8和4T1接种的小鼠模型中,通过将RT与TLR7激动剂DSP-0509组合可以增强抗肿瘤活性。我们发现,需要每周一次(q1w)给药DSP-0509而不是每两周一次(q2w)给药,以在CT26模型中实现优异的抗肿瘤活性。RT/DSP-0509联合治疗组小鼠脾细胞显示肿瘤溶解活性增加,与肿瘤体积呈负相关,如通过铬释放细胞毒性测定法测量的。我们还发现完全治愈的小鼠脾脏中细胞毒性T淋巴细胞(CTL)的水平增加。当通过联合疗法完全治愈的小鼠被CT26细胞重新攻击时,所有小鼠均排斥CT26细胞,但接受Renca细胞.在CD8耗竭时没有观察到这种排斥。此外,联合治疗组的脾效应记忆CD8T细胞水平升高.探讨综合治疗完全治愈的因素,我们分析了完全治愈小鼠的外周血白细胞(PBLs)mRNA。我们发现Havcr2low,Cd274低,Cd80高,和Il6low是联合治疗完全反应的预测特征。肿瘤来源的mRNA的分析显示RT和DSP-0509的组合强烈增加抗肿瘤效应分子(包括Gzmb和Il12)的表达。
    结论:这些数据表明TLR7激动剂,当通过上调CTL活性和效应分子的基因表达与RT组合使用时,DSP-0509可以是有希望的伴随物。这种组合可以是临床试验中预期的新的放射性免疫治疗策略。
    BACKGROUND: TLR7 is a key player in the antiviral immunity. TLR7 signaling activates antigen-presenting cells including DCs and macrophages. This activation results in the adaptive immunity including T cells and B cells. Therefore, TLR7 is an important molecule of the immune system. Based on these observations, TLR7 agonists considered to become a therapy weaponize the immune system against cancer. Radiation therapy (RT) is one of the standard cancer therapies and is reported to modulate the tumor immune response. In this study, we aimed to investigate the anti-tumor activity in combination of TLR7 agonist, DSP-0509, with RT and underlying mechanism.
    RESULTS: We showed that anti-tumor activity is enhanced by combining RT with the TLR7 agonist DSP-0509 in the CT26, LM8, and 4T1 inoculated mice models. We found that once- weekly (q1w) dosing of DSP-0509 rather than biweekly (q2w) dosing is needed to achieve superior anti-tumor activities in CT26 model. Spleen cells from the mice in RT/DSP-0509 combination treatment group showed increased tumor lytic activity, inversely correlated with tumor volume, as measured by the chromium-release cytotoxicity assay. We also found the level of cytotoxic T lymphocytes (CTLs) increased in the spleens of completely cured mice. When the mice completely cured by combination therapy were re-challenged with CT26 cells, all mice rejected CT26 cells but accepted Renca cells. This rejection was not observed with CD8 depletion. Furthermore, levels of splenic effector memory CD8 T cells were increased in the combination therapy group. To explore the factors responsible for complete cure by combination therapy, we analyzed peripheral blood leukocytes (PBLs) mRNA from completely cured mice. We found that Havcr2low, Cd274low, Cd80high, and Il6low were a predictive signature for the complete response to combination therapy. An analysis of tumor-derived mRNA showed that combination of RT and DSP-0509 strongly increased the expression of anti-tumor effector molecules including Gzmb and Il12.
    CONCLUSIONS: These data suggest that TLR7 agonist, DSP-0509, can be a promising concomitant when used in combination with RT by upregulating CTLs activity and gene expression of effector molecules. This combination can be an expecting new radio-immunotherapeutic strategy in clinical trials.
    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:放射疗法和免疫疗法(免疫放射疗法)的组合已越来越多地用于治疗多种癌症。然而,一些肿瘤对免疫放射疗法有抵抗力。我们先前已经表明,在巨噬细胞上表达的MER原癌基因酪氨酸激酶(MerTK)介导了对免疫治疗的抗性。因此,我们寻求开发可以减轻MerTK负面影响的疗法。我们设计并开发了MerTK特异性反义寡核苷酸(ASO),并表征了其在小鼠中引发抗肿瘤免疫反应的作用。
    方法:将344SQR细胞注射到8-12周龄雌性129sv/ev小鼠第0天的右腿和第4天的左腿中,以建立原发性和继发性肿瘤,分别。在第8、9和10天对原发性肿瘤给予12Gy剂量的辐射。小鼠从肿瘤植入后第1天开始接受抗PD-1、抗CTLA-4或/和MerTKASO。通过流式细胞术评估肿瘤微环境的组成和肿瘤中巨噬细胞上的MerTK水平。用NanoString研究免疫相关基因的表达。最后,在组织学上评估了MerTKASO对眼睛结构的影响。
    结果:值得注意的是,在XRT+抗PD1和XRT+抗CTLA4中加入MerTKASO显著减缓了原发性和继发性肿瘤的生长,并显著延长了生存期.ASO显著降低肿瘤相关巨噬细胞(TAMs)中MerTK的表达,将其表型从M2重新编程为M1。此外,当与XRT+抗CTLA4组合时,MerTKASO增加颗粒酶B+CD8+T细胞在继发性肿瘤中的百分比。NanoString结果表明,MerTKASO可有利地调节免疫相关基因,以促进继发性肿瘤中的抗肿瘤免疫反应。重要的是,眼组织的组织学分析表明,与小分子不同,MerTKASO在眼睛中没有产生任何可检测的病理。
    结论:MerTKASO可以显著下调TAMs上MerTK的表达,从而促进抗肿瘤免疫反应。MerTKASO与免疫放射疗法的组合可以安全且显着地减缓肿瘤生长并提高生存率。
    BACKGROUND: The combination of radiotherapy and immunotherapy (immunoradiotherapy) has been increasingly used for treating a wide range of cancers. However, some tumors are resistant to immunoradiotherapy. We have previously shown that MER proto-oncogene tyrosine kinase (MerTK) expressed on macrophages mediates resistance to immunoradiotherapy. We therefore sought to develop therapeutics that can mitigate the negative impact of MerTK. We designed and developed a MerTK specific antisense oligonucleotide (ASO) and characterized its effects on eliciting an anti-tumor immune response in mice.
    METHODS: 344SQR cells were injected into the right legs on day 0 and the left legs on day 4 of 8-12 weeks old female 129sv/ev mice to establish primary and secondary tumors, respectively. Radiation at a dose of 12 Gy was given to the primary tumors on days 8, 9, and 10. Mice received either anti-PD-1, anti-CTLA-4 or/and MerTK ASO starting from day 1 post tumor implantation. The composition of the tumor microenvironment and the level of MerTK on macrophages in the tumor were evaluted by flow cytometry. The expression of immune-related genes was investigated with NanoString. Lastly, the impact of MerTK ASO on the structure of the eye was histologically evaluated.
    RESULTS: Remarkably, the addition of MerTK ASO to XRT+anti-PD1 and XRT+anti-CTLA4 profoundly slowed the growth of both primary and secondary tumors and significantly extended survival. The ASO significantly reduced the expression of MerTK in tumor-associated macrophages (TAMs), reprograming their phenotype from M2 to M1. In addition, MerTK ASO increased the percentage of Granzyme B+ CD8+ T cells in the secondary tumors when combined with XRT+anti-CTLA4. NanoString results demonstrated that the MerTK ASO favorably modulated immune-related genes for promoting antitumor immune response in secondary tumors. Importantly, histological analysis of eye tissues demonstrated that unlike small molecules, the MerTK ASO did not produce any detectable pathology in the eyes.
    CONCLUSIONS: The MerTK ASO can significantly downregulate the expression of MerTK on TAMs, thereby promoting antitumor immune response. The combination of MerTK ASO with immunoradiotherapy can safely and significantly slow tumor growth and improve survival.
    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
    食管癌通常预后不良。鉴于肿瘤免疫疗法的重大突破,越来越多的临床研究表明,放疗和免疫检查点抑制剂(ICIs)联合治疗食管癌可能具有协同作用和良好的预后.从2021年到现在,食管癌的免疫放射疗法(iRT)的临床研究得到了极大的发展。然而,缺乏对食管癌综合治疗的疗效和毒性的总结,以指导临床治疗。对于这篇评论,我们整合最新数据,分析和评估iRT治疗食管癌的疗效和安全性.此外,我们讨论了更好的预测生物标志物,针对特定人群的治疗选择,以及其他挑战,以确定未来研究设计的方向。
    Esophageal cancer usually has a poor prognosis. Given the significant breakthrough with tumor immunotherapy, an increasing number of clinical studies have demonstrated that the combination of radiotherapy and immune checkpoint inhibitors (ICIs) may have a synergistic effect and good outcome in esophageal cancer. Clinical studies of immunoradiotherapy (iRT) for esophageal cancer have proliferated enormously from 2021 to the present. However, a summary of the efficacy and toxicity of combined therapy to guide esophageal cancer treatment in clinical practice is lacking. For this review, we integrate the latest data to analyze and assess the efficacy and safety of iRT for esophageal cancer. In addition, we discuss better predictive biomarkers, therapeutic options for specific populations, and other challenges to identify directions for future research design.
    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
    非小细胞肺癌(NSCLC)的5年生存率极低,唯一有效的治疗方法是免疫放射疗法(iRT)。这里,我们回顾了2018-2023年iRT治疗非小细胞肺癌(NSCLC)的临床研究进展,以及未来的研究方向.我们首先讨论了iRT的协同机制,体现在三个方面:RT的免疫调节,RT激活的免疫相关途径,和RT相关的免疫敏化。iRT可能包括与免疫检查点抑制剂结合的外部束或立体定向体RT(例如,针对免疫程序性细胞死亡的免疫球蛋白(PD)1/PD配体1或CD8+T淋巴细胞抗原4)或中药药物。关于临床有效性和安全性,iRT可提高NSCLC患者的总体生存率和无进展生存率以及肿瘤控制率,但毒性风险没有显著增加.我们最后讨论了2018-2023年报告的iRT挑战和未来方向。
    Non-small-cell lung cancer (NSCLC) has an extremely low 5-year survival rate, with the only effective treatment being immunoradiotherapy (iRT). Here, we review the progress of clinical research on iRT for non-small-cell lung cancer (NSCLC) over 2018-2023, as well as the future directions. We first discuss the synergistic mechanisms of iRT, reflected in three aspects: immune regulation of RT, RT-activated immune-related pathways, and RT-related immune sensitization. iRT may include either external-beam or stereotactic-body RT combined with either immune checkpoint inhibitors (e.g., immunoglobulins against immune programmed cell death (PD) 1/PD ligand 1 or CD8+ T lymphocyte antigen 4) or traditional Chinese medicine drugs. Regarding clinical effectiveness and safety, iRT increases overall and progression-free survival and tumor control rate among patients with NSCLC but without a considerable increase in toxicity risk. We finally discuss iRT challenges and future directions reported over 2018-2023.
    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
    一些临床前数据表明,放射治疗能够调节癌细胞和肿瘤微环境的内在免疫原性。目的是增加对检查点抑制剂的反应。早期结果显示,放疗后患者检查点抑制剂反应的恢复已经产生了很多关于放射治疗的热情,超出了其在局部疾病控制中的通常作用。评估免疫放射疗法组合的前瞻性临床试验提供了概念证明,放射疗法可以在用检查点抑制剂治疗的患者中诱导肿瘤特异性T免疫应答。然而,这些结果并不总是可重复的,反映了与放射治疗相关的因素的存在,免疫疗法和/或宿主,影响这些组合的功效。抗癌化疗可以通过增强肿瘤免疫原性和调节肿瘤微环境来增强免疫-辐射反应。
    Several preclinical data have suggested the ability of radiation therapy to modulate the intrinsic immunogenicity of cancer cells and the tumor microenvironment, with the aim of increasing responses to checkpoint inhibitors. Early results showing a restoration of checkpoint inhibitors response in patients following irradiation have generated a lot of enthusiasm around radiation therapy beyond its usual role in local disease control. Prospective clinical trials evaluating immunoradiotherapy combinations have provided proof-of-concept that radiation therapy may induce tumor-specific T immune responses in patients treated with checkpoint inhibitors. However, these results are not always reproducible, reflecting the existence of factors related to either radiation therapy, immunotherapy and/or the host, which influence the efficacy of these combinations. Anticancer chemotherapy can play a role in amplifying the immune-radiation response by promoting tumor immunogenicity and modulating the tumor microenvironment.
    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
    软组织肉瘤(STS)是来源于间充质组织的高度异质性恶性肿瘤。先进的STS对当前的抗癌治疗方案反应不佳,中位总生存期不到两年。因此,需要新的更有效的STS治疗方法。越来越多的证据表明,免疫治疗和放疗对恶性肿瘤具有协同治疗作用。此外,免疫放射疗法在各种癌症的临床试验中取得了积极的结果。在这次审查中,我们讨论了免疫放射疗法在癌症治疗中的协同机制以及该联合方案在几种癌症治疗中的应用。此外,我们总结了使用免疫放射疗法治疗STS的现有证据以及目前正在进行的相关临床试验.此外,我们确定了使用免疫放射疗法治疗肉瘤的挑战,并提出了克服这些挑战的方法和预防措施。最后,我们提出了临床研究策略和未来的研究方向,以帮助STS的研究和治疗。
    Soft tissue sarcoma (STS) is a highly heterogeneous malignant tumor derived from mesenchymal tissue. Advanced STS has a poor response to the current anti-cancer therapeutic options, with a median overall survival of less than two years. Thus, new and more effective treatment methods for STS are needed. Increasing evidence has shown that immunotherapy and radiotherapy have synergistic therapeutic effects against malignant tumors. In addition, immunoradiotherapy has yielded positive results in clinical trials for various cancers. In this review, we discuss the synergistic mechanism of immunoradiotherapy in cancer treatment and the application of this combined regimen for the treatment of several cancers. In addition, we summarize the existing evidence on the use of immunoradiotherapy for the treatment of STS and the relevant clinical trials that are currently ongoing. Furthermore, we identify challenges in the use of immunoradiotherapy for the treatment of sarcomas and propose methods and precautions for overcoming these challenges. Lastly, we propose clinical research strategies and future research directions to help in the research and treatment of STS.
    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
    由放射治疗和免疫检查点阻断组成的免疫放射疗法的疗效依赖于有效促进全身抗肿瘤免疫反应的激活,同时减少有利于免疫抑制的局部因素。我们之前证明了NBTXR3,一种纳米粒子放射增强剂,在鼠抗PD1耐药转移性肺癌模型中显著改善免疫应答。我们假设在抗PD1和第二代抗CTLA4中添加放射性激活的NBTXR3可以提高治疗效果。为了检验这个假设,我们在小鼠右腿和左腿接种344SQR细胞以建立原发性和继发性肿瘤。在第7天对原发性肿瘤瘤内注射NBTXR3纳米颗粒,然后在第8、9和10天注射三次12Gy辐射。继发性肿瘤在第13天和第14天接受两次1Gy放射。向小鼠给予多轮抗PD1、抗CTLA4或非岩藻糖基化的抗CTLA4。肿瘤的免疫谱分析表明,NBTXR3与免疫放射疗法的组合显着上调了多种抗肿瘤免疫途径的活性,并降低了调节性抑制T细胞的丰度。这种组合有效根除了原发性和继发性肿瘤,并将动物存活率提高到75%。值得注意的是,以前用含NBTXR3的治疗方法治疗,存活小鼠表现出持久的抗肿瘤记忆免疫反应。该数据提供了令人信服的证据,证明当与抗PD1和多个检查点(例如第二代抗CTLA4)组合时,NBTXR3与免疫放射疗法方法协同作用,并显示了NBTXR3抗肿瘤免疫调节作用的临床用途的潜力。
    The efficacy of immunoradiotherapy consisting of radiation therapy and immune checkpoint blockade relies on effectively promoting the systemic antitumor immune response\'s activation while simultaneously reducing local factors favoring immune suppression. We previously demonstrated that NBTXR3, a nanoparticle radioenhancer, significantly improved immune responses in a murine anti-PD1-resistant metastatic lung cancer model. We hypothesize that radioactivated-NBTXR3 addition to anti-PD1 and a second-generation anti-CTLA4 could improve treatment effectiveness. To test this hypothesis, we inoculated mice with 344SQR cells in the right and left legs to establish primary and secondary tumors. The primary tumors were intratumorally injected with NBTXR3 nanoparticles on day 7, followed by three fractions of 12 Gy radiation on days 8, 9, and 10. The secondary tumors received two fractions of 1Gy radiation on days 13 and 14. Multiple rounds of anti-PD1, anti-CTLA4 or nonfucosylated anti-CTLA4 were given to the mice. Immune profiling of the tumors revealed that the combination of NBTXR3 with immunoradiotherapy significantly upregulated the activities of a wide range of antitumor immune pathways and reduced the abundance of regulatory suppressor T cells. This combination effectively eradicated the primary and secondary tumors and increased animal survival to 75%. Remarkably, previously treated with NBTXR3-containing treatment, the survivor mice exhibited a long-lasting antitumor memory immune response. This data provides compelling evidence of the efficacy of NBTXR3 to synergize with the immunoradiotherapy approach when combined with an anti-PD1 and multiple checkpoints such as a second generation anti-CTLA4 and show the potential for clinical uses of antitumor immunomodulatory effects of NBTXR3.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    放射疗法可以触发免疫相关的“外野”反应。我们报告了一名接受长期抗PD1(程序性细胞死亡蛋白1)治疗的晚期NSCLC(非小细胞肺癌)患者,该患者在盆腔照射后发生了场外免疫相关关节炎。
    Radiotherapy can trigger immune-related out-of-field \"abscopal\" response. We report a patient with advanced NSCLC (non-small cell lung cancer) receiving long-term anti-PD1 (programmed cell death protein 1) who have developed out-of-field immune-related arthritis following pelvic irradiation.
    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
    放射治疗(RT)是癌症治疗中的重要方式,其中>50%的癌症患者为了治愈或姑息意图而经历RT。在乳腺癌患者中,肺,和食道癌,以及纵隔恶性肿瘤,心脏或血管结构的附带RT剂量与表现为缺血性心脏病的辐射诱发心脏病(RIHD)的发展有关,心肌病,心功能不全,和心力衰竭。尽管在放射治疗方面取得了显著进展,脱靶心脏毒性是不可避免的。心脏偶然暴露于RT的最佳研究病理后果之一是胶原蛋白沉积和纤维化,导致辐射诱导的心肌纤维化(RIMF)的发展。然而,RIMF的发病机制尚不清楚。此外,一旦RIMF发生,目前尚无可用的临床方法逆转RIMF,并且它继续损害长期癌症幸存者的生活质量.因此,越来越需要更多临床相关的临床前模型来阐明参与RIMF发展的分子和细胞机制.这篇综述提供了对研究RIHD的现有临床前模型和RIMF的建议机制的见解。以及可用的多模态治疗和结果。此外,我们总结了RIHD/RIMF有价值的检测方法,以及敏感的放射学和循环生物标志物的临床应用。
    Radiation therapy (RT) is an important modality in cancer treatment with >50% of cancer patients undergoing RT for curative or palliative intent. In patients with breast, lung, and esophageal cancer, as well as mediastinal malignancies, incidental RT dose to heart or vascular structures has been linked to the development of Radiation-Induced Heart Disease (RIHD) which manifests as ischemic heart disease, cardiomyopathy, cardiac dysfunction, and heart failure. Despite the remarkable progress in the delivery of radiotherapy treatment, off-target cardiac toxicities are unavoidable. One of the best-studied pathological consequences of incidental exposure of the heart to RT is collagen deposition and fibrosis, leading to the development of radiation-induced myocardial fibrosis (RIMF). However, the pathogenesis of RIMF is still largely unknown. Moreover, there are no available clinical approaches to reverse RIMF once it occurs and it continues to impair the quality of life of long-term cancer survivors. Hence, there is an increasing need for more clinically relevant preclinical models to elucidate the molecular and cellular mechanisms involved in the development of RIMF. This review offers an insight into the existing preclinical models to study RIHD and the suggested mechanisms of RIMF, as well as available multi-modality treatments and outcomes. Moreover, we summarize the valuable detection methods of RIHD/RIMF, and the clinical use of sensitive radiographic and circulating biomarkers.
    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
    脑转移是常见原发性癌症的毁灭性后遗症(例如,肺,乳房,和皮肤),并且有效的治疗选择有限。以前,全身化疗未能证明对脑转移患者有显著的益处,但是最近几十年来,靶向治疗和最近的免疫检查点抑制剂(ICIs)在临床前和临床研究中取得了有希望的结果.此外,利用放疗(RT)的免疫调节作用与ICIs协同作用是非常有兴趣的。在这里,我们讨论了评估RT剂量和分级对免疫反应影响的研究,早期研究支持RT和ICIs之间的协同相互作用,和正在进行的临床试验评估联合治疗对脑转移患者的益处。
    Brain metastases are a devastating sequela of common primary cancers (e.g., lung, breast, and skin) and have limited effective therapeutic options. Previously, systemic chemotherapy failed to demonstrate significant benefit in patients with brain metastases, but in recent decades, targeted therapies and more recently immune checkpoint inhibitors (ICIs) have yielded promising results in preclinical and clinical studies. Furthermore, there is significant interest in harnessing the immunomodulatory effects of radiotherapy (RT) to synergize with ICIs. Herein, we discuss studies evaluating the impact of RT dose and fractionation on the immune response, early studies supporting the synergistic interaction between RT and ICIs, and ongoing clinical trials assessing the benefit of combination therapy in patients with brain metastases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号