Solid tumor

实体瘤
  • 文章类型: Journal Article
    背景:许多随机对照试验(RCT)已经研究了基于PD-1/PD-L1抑制剂的联合疗法。围绕两种免疫肿瘤学(IO)疗法对癌症患者的潜在累加临床益处的争论仍然存在。
    方法:涵盖了已发表的随机临床试验和灰色来源,这些试验比较了基于抗PD-1/PD-L1的免疫治疗组合与单一疗法在晚期或转移性实体瘤患者中的治疗。主要结果是无进展生存期(PFS),次要结果包括客观反应率(ORR),总生存期(OS)和治疗相关不良事件(TRAEs)。
    结果:我们的分析涵盖了31项研究,包括10,341名患者,涵盖了12种不同的免疫-肿瘤联合治疗方案。在所有患者中,免疫治疗组合显示出增强ORR(OR=1.23[95%CI1.13~1.34])和延长PFS(HR=0.91[95%CI0.87~0.95])的能力.然而,观察到的OS增强(HR=0.96[95%CI0.91-1.01])无显著性。在PD-L1表达阴性的情况下,PFS(HR=0.82[95%CI0.72至0.93])和OS(HR=0.85[95%CI0.73至0.99])方面的更大益处可能特别明显。值得注意的是,尽管任何等级TRAE(OR=1.72[95%CI1.40-2.11])和等级大于或等于3个TRAE(OR=2.01[95%CI1.67-2.43])的风险增加,毒性通常是可控的。
    结论:这项研究表明,与PD-1/PD-L1抑制剂一起加入额外的免疫治疗剂可以提高反应率并降低疾病进展的风险。同时保持可控的毒性。然而,将这些主要临床获益转化为延长总生存期仍存在挑战.
    BACKGROUND: Numerous randomized controlled trials (RCTs) have investigated PD-1/PD-L1 inhibitor-based combination therapies. The debate surrounding the potential additive clinical benefits of combination of two immune-oncology (IO) therapies for cancer patients persists.
    METHODS: Both published and grey sources of randomized clinical trials that compared anti-PD-1/PD-L1-based immunotherapy combinations with monotherapy in patients with advanced or metastatic solid tumors were encompassed. The primary outcome was progression-free survival (PFS), and secondary outcomes included objective response rate (ORR), overall survival (OS) and treatment-related adverse events (TRAEs).
    RESULTS: Our analysis encompassed 31 studies comprising 10,341 patients, which covered 12 distinct immune-oncology combination regimens. Across all patients, the immunotherapy combinations exhibited the capability to enhance the ORR (OR = 1.23 [95% CI 1.13-1.34]) and extend PFS (HR = 0.91 [95% CI 0.87-0.95]). However, the observed enhancement in OS (HR = 0.96 [95% CI 0.91-1.01]) was of no significance. Greater benefits in terms of PFS (HR = 0.82 [95% CI 0.72 to 0.93]) and OS (HR = 0.85 [95% CI 0.73 to 0.99]) may be particularly pronounced in cases where PD-L1 expression is negative. Notably, despite a heightened risk of any-grade TRAEs (OR = 1.72 [95% CI 1.40-2.11]) and grade greater than or equal to 3 TRAEs (OR = 2.01 [95% CI 1.67-2.43]), toxicity was generally manageable.
    CONCLUSIONS: This study suggests that incorporating an additional immunotherapy agent with PD-1/PD-L1 inhibitors can elevate the response rate and reduce the risk of disease progression, all while maintaining manageable toxicity. However, there remains a challenge in translating these primary clinical benefits into extended overall 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: Systematic Review
    目前,嵌合抗原受体T细胞(CAR-T)和免疫检查点抑制剂(如程序性细胞死亡蛋白-1(PD-1)阻断剂)等疗法对许多癌症患者显示出有希望的结果。然而,在CAR-T疗法成为现成的治疗方法之前,需要取得重大进展,特别是实体瘤和淋巴瘤。在这次审查中,我们系统分析了涉及工程化CAR-T细胞和抗PD-1药物的联合治疗.这种方法旨在克服当前治疗的局限性,并提供潜在的优势,例如增强的肿瘤抑制作用。减轻T细胞耗尽,T细胞激活增强,和最小化的毒性。CAR-T疗法的整合,靶向肿瘤相关抗原,PD-1阻断增强T细胞功能并减轻肿瘤微环境内的免疫抑制。评估联合治疗对各种肿瘤和淋巴瘤的影响,我们根据六种主要的肿瘤相关抗原对它们进行分类:间皮素,二唾液酸神经节苷脂GD-2,CD-19,CD-22,CD-133和CD-30,存在于不同的肿瘤类型。我们评估了疗效,完整和部分响应,以及临床前和临床模型中的无进展生存期。此外,我们讨论了潜在的影响,包括联合免疫疗法的可行性,强调正在进行的研究对优化癌症患者治疗策略和改善预后的重要性。总的来说,我们认为,CAR-T疗法与PD-1阻断联合应用有望成为下一代癌症免疫疗法.
    Currently, therapies such as chimeric antigen receptor-T Cell (CAR-T) and immune checkpoint inhibitors like programmed cell death protein-1 (PD-1) blockers are showing promising results for numerous cancer patients. However, significant advancements are required before CAR-T therapies become readily available as off-the-shelf treatments, particularly for solid tumors and lymphomas. In this review, we have systematically analyzed the combination therapy involving engineered CAR-T cells and anti PD-1 agents. This approach aims at overcoming the limitations of current treatments and offers potential advantages such as enhanced tumor inhibition, alleviated T-cell exhaustion, heightened T-cell activation, and minimized toxicity. The integration of CAR-T therapy, which targets tumor-associated antigens, with PD-1 blockade augments T-cell function and mitigates immune suppression within the tumor microenvironment. To assess the impact of combination therapy on various tumors and lymphomas, we categorized them based on six major tumor-associated antigens: mesothelin, disialoganglioside GD-2, CD-19, CD-22, CD-133, and CD-30, which are present in different tumor types. We evaluated the efficacy, complete and partial responses, and progression-free survival in both pre-clinical and clinical models. Additionally, we discussed potential implications, including the feasibility of combination immunotherapies, emphasizing the importance of ongoing research to optimize treatment strategies and improve outcomes for cancer patients. Overall, we believe combining CAR-T therapy with PD-1 blockade holds promise for the next generation of cancer immunotherapy.
    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
    基因工程的最新进展使修饰自然杀伤(NK)细胞成为可能,以增强其对抗各种癌症的能力。包括实体瘤.本综述讨论了基因工程嵌合抗原受体NK细胞疗法的现状及其治疗实体瘤的潜力。我们探讨NK细胞的固有特性及其在免疫调节和肿瘤监测中的作用。此外,我们研究了用于基因改造NK细胞的策略,安全概况,和潜在的临床应用。我们的研究表明,CAR-NK细胞可以有效地靶向和消退非血液恶性肿瘤,显示增强的抗肿瘤功效。这意味着使用遗传修饰的NK细胞治疗肿瘤的极好前景。值得注意的是,NK细胞表现出低的移植物抗宿主病(GvHD)潜力,很少诱导显著的毒性,使它们成为汽车工程的理想平台。同种异体NK细胞过继转移到患者中进一步强调了NK细胞用于各种应用的多功能性。我们还解决了与基因工程NK细胞疗法的临床翻译相关的挑战和局限性,比如脱靶效应,免疫逃逸机制,和制造可扩展性。我们提供了通过组合疗法和递送优化来克服这些障碍的策略。总的来说,我们相信这篇综述通过阐明潜在的机制,有助于推进NK细胞免疫疗法作为一种有前途的癌症治疗方法,评估临床前和临床证据,解决剩余的挑战。
    Recent advancements in genetic engineering have made it possible to modify Natural Killer (NK) cells to enhance their ability to fight against various cancers, including solid tumors. This comprehensive overview discusses the current status of genetically engineered chimeric antigen receptor NK-cell therapies and their potential for treating solid tumors. We explore the inherent characteristics of NK cells and their role in immune regulation and tumor surveillance. Moreover, we examine the strategies used to genetically engineer NK cells in terms of efficacy, safety profile, and potential clinical applications. Our investigation suggests CAR-NK cells can effectively target and regress non-hematological malignancies, demonstrating enhanced antitumor efficacy. This implies excellent promise for treating tumors using genetically modified NK cells. Notably, NK cells exhibit low graft versus host disease (GvHD) potential and rarely induce significant toxicities, making them an ideal platform for CAR engineering. The adoptive transfer of allogeneic NK cells into patients further emphasizes the versatility of NK cells for various applications. We also address challenges and limitations associated with the clinical translation of genetically engineered NK-cell therapies, such as off-target effects, immune escape mechanisms, and manufacturing scalability. We provide strategies to overcome these obstacles through combination therapies and delivery optimization. Overall, we believe this review contributes to advancing NK-cell-based immunotherapy as a promising approach for cancer treatment by elucidating the underlying mechanisms, evaluating preclinical and clinical evidence, and addressing remaining challenges.
    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
    乳腺癌(BC)异质性是BC肿瘤的关键特征,对肿瘤发生具有重要意义。诊断,和治疗方式。它受肿瘤内在特征和不同肿瘤内区域的肿瘤微环境(TME)组成的影响,进而影响患者体内的癌症进展。在这个迷你评论中,我们将重点介绍在BC中产生癌症异质性的机制,以及它们如何影响对癌症治疗的反应。
    Breast cancer (BC) heterogeneity is a key trait of BC tumors with crucial implications on tumorigenesis, diagnosis, and therapeutic modalities. It is influenced by tumor intrinsic features and by the tumor microenvironment (TME) composition of different intra-tumoral regions, which in turn affect cancer progression within patients. In this mini review, we will highlight the mechanisms that generate cancer heterogeneity in BC and how they affect the responses to cancer therapies.
    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
    目的探讨乐伐替尼治疗不同类型实体瘤的疗效和安全性。方法通过搜索PubMed,WebofScience,科克伦,CNKI,万方等数据库,所有文献都是关于lenvatinib治疗各种实体瘤的临床疗效比较。根据文献的纳入和排除标准,两名参与者筛选了文献,整理数据并对文献进行评估。采用RevMan5.4软件对纳入文献进行Meta分析。结果共纳入12项研究,包括5213名患者。Meta分析显示,就功效而言,lenvatinib组治疗各种实体瘤延长PFS的风险(HR)是对照组的1.91倍(HR=1.91,95%CI:1.58-2.31,P<0.00001),单一靶向药物组OS延长的风险(HR)是单一靶向药物组的1.27倍(HR=1.27,95%CI:1.15-1.40,P<0.00001)。在安全方面,lenvatinib组治疗各种实体瘤发生不良事件的风险高于对照组,尤其是内分泌毒性,肾/尿毒性,血管毒性,肌肉骨骼/结缔组织毒性和代谢/营养毒性。结论Lenvatinib治疗多种实体瘤可以延长患者的OS和PFS,提高临床获益率,改善患者生活质量。同时,有一定的不良事件发生率,临床用药中应给予对症干预。
    UNASSIGNED: The purpose of this study was to investigate the efficacy and safety of lenvatinib in various types of solid tumors.
    UNASSIGNED: By searching PubMed, Web of Science, Cochrane, CNKI, Wanfang and other databases, all the literatures about the comparison of clinical efficacy of lenvatinib in the treatment of various solid tumors. According to the criteria of inclusion and exclusion of literature, two participants screened the literature, collated the data and evaluated the literature. RevMan 5.4 software was used for meta-analysis of the included literatures.
    UNASSIGNED: A total of 12 studies were included, including 5213 patients. Meta-analysis showed that, in terms of efficacy, the risk (HR) of prolonging PFS in the treatment of various solid tumors in the lenvatinib group was 1.91 times that in the control group (HR = 1.91, 95% CI: 1.58-2.31, p < 0.00001), and the risk (HR) of prolonging OS was 1.27 times that in the single targeted drug group (HR = 1.27, 95% CI: 1.15-1.40, p < 0.00001). In terms of safety, the risk of adverse events in the treatment of various solid tumors in the lenvatinib group was higher than that in the control group, especially in Endocrine Toxicities, Renal/Urinary Toxicities, Vascular Toxicities, Musculoskeletal/a Connective Tissue Toxicities and Metabolism/Nutrition Toxicities.
    UNASSIGNED: Lenvatinib in various solid tumors can prolong OS and disease PFS of patients, improve the clinical benefit rate and improve the quality of life of patients. At the same time, there is a certain incidence of adverse events, and symptomatic intervention should be given in clinical medication.
    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
    肿瘤浸润淋巴细胞(TIL)的过继性细胞疗法在转移性黑色素瘤患者中表现出持久的临床反应,最近关于TIL治疗的第一个III期试验的阳性结果证实了这一点。作为一种苛刻且后勤复杂的治疗方法,需要广泛的临床前和临床努力,以通过识别反应的预测生物标志物来优化患者选择。这篇综述旨在全面总结当前关于肿瘤相关因素(如突变负担,新抗原负荷,免疫浸润,致癌驱动基因的状态,和表观遗传修饰),患者特征(包括疾病负担和位置,基线细胞因子和乳酸脱氢酶血清水平,人类白细胞抗原单倍型,或事先接触过免疫检查点抑制剂和其他抗癌疗法),转移的T细胞的表型特征(主要是总细胞计数,CD8:CD4比值,离体培养时间,耗尽标记的表达,共刺激信号,抗肿瘤反应性,和靶肿瘤相关抗原的范围),和其他治疗相关因素(如淋巴消耗化疗和输注白细胞介素-2后)。
    Adoptive cell therapy with tumor-infiltrating lymphocytes (TIL) has demonstrated durable clinical responses in patients with metastatic melanoma, substantiated by recent positive results of the first phase III trial on TIL therapy. Being a demanding and logistically complex treatment, extensive preclinical and clinical effort is required to optimize patient selection by identifying predictive biomarkers of response. This review aims to comprehensively summarize the current evidence regarding the potential impact of tumor-related factors (such as mutational burden, neoantigen load, immune infiltration, status of oncogenic driver genes, and epigenetic modifications), patient characteristics (including disease burden and location, baseline cytokines and lactate dehydrogenase serum levels, human leucocyte antigen haplotype, or prior exposure to immune checkpoint inhibitors and other anticancer therapies), phenotypic features of the transferred T cells (mainly the total cell count, CD8:CD4 ratio, ex vivo culture time, expression of exhaustion markers, costimulatory signals, antitumor reactivity, and scope of target tumor-associated antigens), and other treatment-related factors (such as lymphodepleting chemotherapy and postinfusion administration of interleukin-2).
    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
    实体瘤的骨髓转移(BMM)是指一组起源于非造血恶性肿瘤细胞通过复杂的转移模式侵入骨髓(BM)的疾病。如果BMM识别延迟,该疾病将迅速发展为BM的播散性致癌作用,表现为一系列血液系统疾病和微血管病性溶血性贫血,导致严重的危及生命的情况。虽然实体瘤BMM的研究越来越受到重视,研究仍然有限,大多数描述来自病例报告。目前,临床医生对BMM的认识不足,BMM的发生往往不能早期识别或有效治疗,导致高死亡率。在这篇文章中,我们回顾了流行病学,分子机制,临床诊断,治疗,实体瘤BMM的预后。
    Bone marrow metastasis (BMM) of solid tumors refers to a group of diseases that originate from non-hematopoietic malignant tumor cells invading the bone marrow (BM) through complex metastatic patterns. If BMM identification is delayed, the disease will rapidly develop into disseminated carcinogenesis of the BM, which manifests as a series of hematological disorders and microangiopathic hemolytic anemia, leading to serious life-threatening conditions. Although the study of solid tumor BMM is receiving increasing attention, study remains limited, and most descriptions are derived from case reports. Currently, clinicians have insufficient understanding of BMM, and BMM occurrence is often not recognized early or treated effectively, resulting in high mortality rates. In this article, we review the epidemiology, molecular mechanisms, clinical diagnosis, treatment, and prognosis of solid tumor BMM.
    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
    由于其对治疗的抗性,肺癌在世界范围内具有高发病率。因此,需要新的治疗方案来改善肺癌治疗的结果。这项研究旨在评估溶瘤病毒(OVs)作为一种新型癌症治疗方法的有效性。在这项研究中,回顾了PubMed和Scopus在1994年至2022年发表的158篇关于OVs在肺癌治疗中的有效性的文章。研究了八类OVs的溶瘤特性及其与治疗方案的相互作用。OVs可以作为一种有前途的免疫疗法,因为它们在不同类型的癌细胞中选择性地繁殖,导致肿瘤细胞裂解并引发有效的免疫反应。
    为了找到治疗肺癌的方法,已经进行了大量的研究。研究的方法之一是使用一种称为溶瘤病毒(OV)的病毒治疗癌症。由于肿瘤具有独特的性质,OV倾向于与它们结合并激活免疫细胞以杀死它们。本文回顾了OVs与其他常见癌症治疗方法的结合,以提高其有效性。引起更少的反应,并带来更好的结果。
    Lung cancer has a high morbidity rate worldwide due to its resistance to therapy. So new treatment options are needed to improve the outcomes of lung cancer treatment. This study aimed to evaluate the effectiveness of oncolytic viruses (OVs) as a new type of cancer treatment. In this study, 158 articles from PubMed and Scopus from 1994 to 2022 were reviewed on the effectiveness of OVs in the treatment of lung cancer. The oncolytic properties of eight categories of OVs and their interactions with treatment options were investigated. OVs can be applied as a promising immunotherapy option, as they are reproduced selectively in different types of cancer cells, cause tumor cell lysis and trigger efficient immune responses.
    A lot of research has been done to find a cure for lung cancer. Among the methods investigated is the treatment of cancer using a type of virus called an oncolytic virus (OV). Since tumors have unique properties, OVs tend to bind to them and activate immune cells to kill them. This article reviews the combination of OVs with other common cancer treatments which improves their effectiveness, causes fewer reactions and brings better results.
    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:淋巴细胞激活基因3(LAG3)是最近发现的免疫检查点分子,与免疫抑制和不同类型实体瘤中癌症的发展有关。本研究旨在评估LAG3的预后重要性及其在实体瘤免疫系统中的作用。
    方法:使用Pubmed,EMBASE,和Cochrane图书馆数据库,以确定探索LAG3对生存结局影响的相关研究。计算合并风险比(HR)及其95%置信区间(CI)以评估LAG3的预后价值。之后,进行亚组分析和敏感性分析。泛癌分析调查了LAG3表达与遗传改变之间的可能关系,RNA甲基化修饰相关基因,基因组不稳定性,免疫检查点基因,和免疫细胞的浸润。
    结果:本分析共纳入43项研究,共7,118名患者。LAG3的高表达与较差的总生存期相关(HR=1.10,95%CI1.01-1.19,P=0.023)。但非无病生存(HR=1.41,95%CI0.96-2.07,P=0.078),无进展生存期(HR=1.12,95%CI0.90-1.39,P=0.317)或无复发生存期(HR=0.98,95%CI0.81-1.19,P=0.871).亚组分析显示LAG3在不同实体瘤中可能发挥不同的预后作用。LAG3表达与所有癌症中的免疫细胞浸润和免疫检查点基因呈正相关。还发现LAG3表达与微卫星不稳定性(MSI)有关,拷贝数变异(CNV),简单核苷变异(SNV),肿瘤突变负荷(TMB),和新抗原在各种类型的癌症。
    结论:LAG3表达升高与诊断为实体癌的患者预后较差有关。LAG3在不同类型的实体肿瘤中可能发挥不同的预后作用。鉴于其在癌症免疫和肿瘤发生中的大量参与,LAG3作为一种有前途的预后生物标志物和免疫疗法的潜在靶标而受到关注。
    BACKGROUND: Lymphocyte-activation gene 3 (LAG3) is a recently discovered immune checkpoint molecule that has been linked to immunosuppression and the advancement of cancer in different types of solid tumors. This study aimed to evaluate the prognostic importance of LAG3 and its role in the immune system within solid tumors.
    METHODS: Extensive literature searches were conducted using the Pubmed, EMBASE, and Cochrane Library databases to identify relevant studies exploring the effect of LAG3 on survival outcomes. Pooled hazard ratios (HRs) with its 95% confidence intervals (CIs) were calculated to evaluate the prognostic values of LAG3. Afterwards, subgroup analysis and sensitivity analysis were conducted. Pan-cancer analysis investigated the possible relationships between LAG3 expression and genetic alterations, RNA methylation modification-related genes, genomic instability, immune checkpoint genes, and infiltration of immune cells.
    RESULTS: A total of 43 studies with 7,118 patients were included in this analysis. Higher expression of LAG3 was associated with worse overall survival (HR = 1.10, 95% CI 1.01-1.19, P = 0.023), but not disease-free survival (HR = 1.41, 95% CI 0.96-2.07, P = 0.078), progression-free survival (HR = 1.12, 95% CI 0.90-1.39, P = 0.317) or recurrence-free survival (HR = 0.98, 95% CI 0.81-1.19, P = 0.871). Subgroup analysis showed that LAG3 might play different prognostic roles in different solid tumors. LAG3 expression was positively associated with immune cell infiltration and immune checkpoint genes in all of the cancers included. LAG3 expression was also found to be associated with microsatellite instability (MSI), copy number variation (CNV), simple nucleoside variation (SNV), tumor mutation burden (TMB), and neoantigen in various types of cancers.
    CONCLUSIONS: Elevated expression of LAG3 is linked to poorer prognosis among patients diagnosed with solid cancers. LAG3 might play varying prognostic roles in different types of solid tumors. Given its substantial involvement in cancer immunity and tumorigenesis, LAG3 has garnered attention as a promising prognostic biomarker and a potential target for immunotherapy.
    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
    T细胞增殖调节因子(Tcprs),它们是促进T细胞功能的正调节因子,为改善T细胞功能的疗法的发展做出了巨大贡献。CAR(嵌合抗原受体)-T细胞疗法,一种靶向肿瘤细胞并增强免疫致死性的过继性细胞转移疗法,导致血液肿瘤的治疗取得重大进展。然而,CAR-T在实体瘤治疗中的应用仍然有限。因此,在这次审查中,我们专注于Tcprs用于实体瘤治疗和预后预测的发展。我们总结了靶向不同Tcpr以增强T细胞增殖以及激活和抑制癌症进展的潜在策略。从而提高CAR-T的抗肿瘤活性和持久性。总之,我们提出了通过表达不同的Tcpr来增强CAR-T细胞的方法,这可能导致新一代细胞疗法的发展。
    T cell proliferation regulators (Tcprs), which are positive regulators that promote T cell function, have made great contributions to the development of therapies to improve T cell function. CAR (chimeric antigen receptor) -T cell therapy, a type of adoptive cell transfer therapy that targets tumor cells and enhances immune lethality, has led to significant progress in the treatment of hematologic tumors. However, the applications of CAR-T in solid tumor treatment remain limited. Therefore, in this review, we focus on the development of Tcprs for solid tumor therapy and prognostic prediction. We summarize potential strategies for targeting different Tcprs to enhance T cell proliferation and activation and inhibition of cancer progression, thereby improving the antitumor activity and persistence of CAR-T. In summary, we propose means of enhancing CAR-T cells by expressing different Tcprs, which may lead to the development of a new generation of cell therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号