target therapy

靶向治疗
  • 文章类型: Journal Article
    热休克蛋白是重要的分子伴侣,在稳定蛋白质结构中起着至关重要的作用。促进受损蛋白质的修复或降解,维持蛋白质稳定和细胞功能。大量研究表明,热休克蛋白在癌症中高表达,与肿瘤发生和发展密切相关。“癌症的标志”是癌症生物学的核心特征,它们共同定义了细胞从正常状态过渡到肿瘤生长状态时获得的一系列功能特征,包括持续增殖信号,逃避生长抑制因子,抵抗细胞死亡,启用复制永生,血管生成的诱导,以及侵袭和转移的激活。热休克蛋白通过激活或抑制各种信号通路在调节癌症标志中的关键作用已得到充分证明。因此,本文从癌症标志的角度概述了热休克蛋白在重要生物过程中的作用,并总结了靶向热休克蛋白调节各种癌症标志的小分子抑制剂。此外,我们进一步讨论了涉及热休克蛋白和有前景的双靶点抑制剂的联合治疗策略,以突出靶向热休克蛋白治疗癌症的潜力.总之,这篇综述强调了靶向热休克蛋白如何调节癌症的标志,这将为更好地阐明和理解热休克蛋白在肿瘤学中的作用以及癌症发生和发展的机制提供有价值的信息,并有助于开发更有效,毒性更低的新型抗癌药物。
    Heat shock proteins are essential molecular chaperones that play crucial roles in stabilizing protein structures, facilitating the repair or degradation of damaged proteins, and maintaining proteostasis and cellular functions. Extensive research has demonstrated that heat shock proteins are highly expressed in cancers and closely associated with tumorigenesis and progression. The \"Hallmarks of Cancer\" are the core features of cancer biology that collectively define a series of functional characteristics acquired by cells as they transition from a normal state to a state of tumor growth, including sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, enabled replicative immortality, the induction of angiogenesis, and the activation of invasion and metastasis. The pivotal roles of heat shock proteins in modulating the hallmarks of cancer through the activation or inhibition of various signaling pathways has been well documented. Therefore, this review provides an overview of the roles of heat shock proteins in vital biological processes from the perspective of the hallmarks of cancer and summarizes the small-molecule inhibitors that target heat shock proteins to regulate various cancer hallmarks. Moreover, we further discuss combination therapy strategies involving heat shock proteins and promising dual-target inhibitors to highlight the potential of targeting heat shock proteins for cancer treatment. In summary, this review highlights how targeting heat shock proteins could regulate the hallmarks of cancer, which will provide valuable information to better elucidate and understand the roles of heat shock proteins in oncology and the mechanisms of cancer occurrence and development and aid in the development of more efficacious and less toxic novel anticancer agents.
    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
    免疫毒素(IT)是专门设计用于靶向治疗的治疗剂,特别是在癌症治疗中。它们由与强效细胞毒性剂连接的单克隆抗体或抗体片段组成,如细菌或植物衍生的毒素,如白喉毒素,蓖麻毒素,或者假单胞菌外毒素.单克隆抗体成分与靶细胞表面表达的抗原特异性结合,促进信息技术的内部化。一旦进入牢房,细胞毒性剂被释放,破坏基本的细胞过程并导致细胞死亡。这种靶向方法使对健康组织的损伤最小化,同时有效地消除患病细胞。IT的生产涉及两种主要方法:重组融合和化学缀合。在重组融合中,基因工程被用来创造一种融合抗体和毒素的融合蛋白,确保精确控制他们的比例和功能。在化学偶联中,预先存在的抗体与毒素化学连接,允许在组合不同的抗体和细胞毒性剂方面具有更大的灵活性。每种方法都有其优点和挑战,影响特异性,生产复杂性,以及由此产生的ITs的治疗潜力。随着研究的进展,ITs不仅在肿瘤学中而且在治疗其他疾病方面继续显示出希望,包括炎症和动脉粥样硬化.ITs的精确靶向和有效作用使其成为开发新治疗策略的有价值的工具。
    Immunotoxins (ITs) are specialized therapeutic agents designed for targeted treatment, particularly in cancer therapy. They consist of a monoclonal antibody or antibody fragment linked to a potent cytotoxic agent, such as bacterial- or plant-derived toxins like diphtheria toxin, ricin, or pseudomonas exotoxin. The monoclonal antibody component specifically binds to antigens expressed on the surface of target cells, facilitating the internalization of the IT. Once inside the cell, the cytotoxic agent is released, disrupting essential cellular processes and leading to cell death. This targeted approach minimizes damage to healthy tissues while effectively eliminating diseased cells. The production of ITs involves two primary methods: recombinant fusion and chemical conjugation. In recombinant fusion, genetic engineering is used to create a fusion protein that combines the antibody and toxin, ensuring precise control over their ratio and functionality. In chemical conjugation, pre-existing antibodies are chemically linked to toxins, allowing for greater flexibility in combining different antibodies and cytotoxic agents. Each method has its advantages and challenges, influencing the specificity, production complexity, and therapeutic potential of the resulting ITs. As research advances, ITs continue to show promise not only in oncology but also in treating other diseases, including inflammatory conditions and atherosclerosis. The precise targeting and potent effects of ITs make them a valuable tool in the development of new therapeutic strategies.
    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)长期以来一直是肿瘤学中的一个巨大挑战,部分原因是它们的稀有性和多样性,这使得大规模研究变得复杂,并减缓了新疗法的出现。传统上以蒽环类药物为基础的化疗为基础,在过去的二十年中,STS治疗的格局没有发生重大变化。然而,最近的研究进展开始描绘一幅更有希望的画面。利用先进的分子谱分析,研究人员现在正在针对肿瘤独特的基因组成定制治疗方法,有针对性的疗法显示出希望。诸如用于NTRK重排肉瘤的NTRK抑制剂和用于韧带样肿瘤的γ-分泌酶抑制剂等创新正在改变临床实践。免疫疗法的兴起,包括LAG-3抑制剂和靶向TGF-β和PD-L1的双功能蛋白等新型药物,为治疗提供了新的途径,特别是与化疗等传统疗法结合使用时。同时,对特定肉瘤亚型的表观遗传学治疗的批准预示着基于组织学特异性的新一波策略,这可能会导致更个性化和有效的护理。虽然挑战依然存在,STS治疗领域正在发展,由对疾病的生物学基础的更深入的了解和对创新研究方法的承诺所驱动。
    Soft tissue sarcomas (STS) have long been a formidable challenge in oncology, partly because of their rarity and diversity, which complicates large-scale studies and slows the advent of new treatments. Traditionally anchored by anthracycline-based chemotherapy, the landscape of STS treatment hasn\'t shifted dramatically in the past twenty years. However, recent strides in research are starting to paint a more hopeful picture. Leveraging advanced molecular profiling, researchers are now tailoring treatments to the unique genetic makeup of tumors, with targeted therapies showing promise. Innovations such as NTRK inhibitors for NTRK-rearranged sarcomas and gamma-secretase inhibitors for desmoid tumors are changing clinical practices. The rise of immunotherapy, including novel agents like LAG-3 inhibitors and bifunctional proteins that target both TGF-β and PD-L1, offers new avenues for treatment, particularly when combined with traditional therapies like chemotherapy. Meanwhile, the approval of epigenetic treatments for specific sarcoma subtypes heralds a new wave of strategy based on histological specificity, which could lead to more personalized and effective care. While challenges remain, the field of STS treatment is evolving, driven by a deeper understanding of the disease\'s biological underpinnings and a commitment to innovative research approaches.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    具有靶基因突变的患者经常从靶疗法获得显著的临床益处。然而,患者之间突变丰度水平的差异导致不同的生存获益,甚至在具有相同靶基因突变的患者中。目前,缺乏合理和可解释的模型来评估治疗失败的风险。在这项研究中,我们调查了导致药物敏感性变化的潜在耦合因素,并建立了一个可统计解释的框架,名为SAFE-MIL,用于风险估计。我们首先从探索患者阳性判断值的最佳分组的角度为每个患者构建有效性标签,并将患者分为600和1,000组,分别,基于多实例学习(MIL)。基于此框架的Hosmer-Lemeshow测试,进一步设计了一种新颖的可解释的损失函数。通过将多实例学习与Hosmer-Lemeshow测试集成,SAFE-MIL能够准确估计不同患者队列中药物治疗失败的风险,并为同时评估风险分层提供最佳阈值。我们进行了一项全面的病例研究,涉及457例用EGFR酪氨酸激酶抑制剂治疗的EGFR突变的非小细胞肺癌患者。结果表明,SAFE-MIL优于传统回归方法,具有更高的准确性,可以准确评估患者的风险分层。这强调了其在提供统计可解释性的同时准确捕获患者间风险差异的能力。SAFE-MIL能够有效地指导有关靶向治疗中药物使用的临床决策,并为其他患者分层问题提供可解释的计算框架。SAFE-MIL框架已证明其在捕获患者间风险差异和提供统计可解释性方面的有效性。其性能优于传统回归方法,可有效指导临床靶向治疗药物的使用决策。SAFE-MIL提供了一个有价值的可解释的计算框架,可以应用于其他患者分层问题,提高个性化医疗风险评估的准确性。SAFE-MIL的源代码可在https://github.com/Nevermore233/SAFE-MIL上进一步探索和应用。
    Patients with the target gene mutation frequently derive significant clinical benefits from target therapy. However, differences in the abundance level of mutations among patients resulted in varying survival benefits, even among patients with the same target gene mutations. Currently, there is a lack of rational and interpretable models to assess the risk of treatment failure. In this study, we investigated the underlying coupled factors contributing to variations in medication sensitivity and established a statistically interpretable framework, named SAFE-MIL, for risk estimation. We first constructed an effectiveness label for each patient from the perspective of exploring the optimal grouping of patients\' positive judgment values and sampled patients into 600 and 1,000 groups, respectively, based on multi-instance learning (MIL). A novel and interpretable loss function was further designed based on the Hosmer-Lemeshow test for this framework. By integrating multi-instance learning with the Hosmer-Lemeshow test, SAFE-MIL is capable of accurately estimating the risk of drug treatment failure across diverse patient cohorts and providing the optimal threshold for assessing the risk stratification simultaneously. We conducted a comprehensive case study involving 457 non-small cell lung cancer patients with EGFR mutations treated with EGFR tyrosine kinase inhibitors. Results demonstrate that SAFE-MIL outperforms traditional regression methods with higher accuracy and can accurately assess patients\' risk stratification. This underscores its ability to accurately capture inter-patient variability in risk while providing statistical interpretability. SAFE-MIL is able to effectively guide clinical decision-making regarding the use of drugs in targeted therapy and provides an interpretable computational framework for other patient stratification problems. The SAFE-MIL framework has proven its effectiveness in capturing inter-patient variability in risk and providing statistical interpretability. It outperforms traditional regression methods and can effectively guide clinical decision-making in the use of drugs for targeted therapy. SAFE-MIL offers a valuable interpretable computational framework that can be applied to other patient stratification problems, enhancing the precision of risk assessment in personalized medicine. The source code for SAFE-MIL is available for further exploration and application at https://github.com/Nevermore233/SAFE-MIL.
    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
    系统评估安洛替尼靶向治疗晚期消化系统肿瘤(DSN)患者的疗效和安全性。
    临床试验摘自PubMed,Cochrane图书馆,WebofScience,Embase,截至2023年10月的中国国家知识基础设施(CNKI)和万方数据库。成果措施,包括治疗功效,生活质量(QOL)和不良事件,进行了提取和评估。
    二十次试验,包括1,613名晚期DSN患者,包括在内。结果表明,与单独的常规治疗相比,安洛替尼靶向治疗与常规治疗相结合可显着提高患者的6个月总生存期(OS,OR=1.76,CI=1.53至2.02,P<0.00001),总体反应(ORR,OR=1.76,CI=1.53至2.02,P<0.00001)和疾病控制率(DCR,OR=1.51,95%CI=1.25~1.84,P<0.0001)。此外,接受联合治疗的组高血压发生率更高(P<0.00001),蛋白尿(P<0.00001),疲劳(P<0.00001),腹泻(P<0.00001),高甘油三酯血症(P=0.02),丙氨酸氨基转移酶(ALT)增加(P=0.004),天冬氨酸转氨酶(AST)升高(P=0.006),厌食症(P<0.00001),体重减轻(P=0.002),腹痛(P=0.0006),甲状腺功能减退(P=0.02),QT间期延长(P=0.04)。其他不良事件分析,如胃肠道反应,白细胞减少症,和中性粒细胞减少症,差异不显著(P>0.05)。
    安洛替尼靶向治疗与常规治疗相结合,对DSNs治疗比单独使用常规治疗更有效。然而,这种联合治疗可能导致更高的高血压发病率,蛋白尿和手足综合征。因此,治疗前应考虑获益和风险。
    UNASSIGNED: To systematically evaluate the efficacy and safety of anlotinib targeted therapy for the treatment of patients with advanced digestive system neoplasms (DSNs).
    UNASSIGNED: Clinical trials were extracted from PubMed, the Cochrane Library, Web of Science, Embase, China National Knowledge Infrastructure (CNKI) and the Wanfang database up to October 2023. Outcome measures, including therapeutic efficacy, quality of life (QOL) and adverse events, were extracted and evaluated.
    UNASSIGNED: Twenty trials, including 1,613 advanced DSNs patients, were included. The results indicated that, compared with conventional treatment alone, the combination of anlotinib targeted therapy with conventional treatment significantly improved the patients\' 6-months overall survival (OS, OR=1.76, CI=1.53 to 2.02, P<0.00001), overall response (ORR, OR=1.76, CI=1.53 to 2.02, P<0.00001) and disease control rate (DCR, OR=1.51, 95% CI=1.25 to 1.84, P<0.0001). Moreover, the group that received the combined therapy had higher rates of hypertension (P<0.00001), proteinuria (P<0.00001), fatigue (P<0.00001), diarrhea (P<0.00001), hypertriglyceridemia (P=0.02), alanine aminotransfease (ALT)increased (P=0.004), aspartate transaminase (AST) increased (P=0.006), anorexia (P<0.00001), weight loss (P=0.002), abdominal pain (P=0.0006), hypothyroidism (P=0.02), prolonged QT interval (P=0.04). Analyses of other adverse events, such as gastrointestinal reaction, leukopenia, and neutropenia, did not reveal significant differences (P>0.05).
    UNASSIGNED: The combination of anlotinib targeted therapy and conventional treatment is more effective for DSNs treatment than conventional treatment alone. However, this combined treatment could lead to greater rates of hypertension, albuminuria and hand-foot syndrome. Therefore, the benefits and risks should be considered before treatment.
    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RNA的下一代测序(RNA-seq)代表了检测非小细胞肺癌(NSCLC)基因融合的黄金标准。尽管如此,RNA的不稳定性使得组织样本的管理极其复杂,导致大量测试失败,丢失数据或需要切换到其他技术。在本研究中,我们分析了来自NSCLC患者的140份肿瘤组织样本的分析前变量,以检测增加RNA-seq成功机会的特征.我们发现分析的成功率与RNA浓度和片段化指数呈正相关。有趣的是,与手术标本和细胞块相比,小活检是更合适的样本.在手术标本中,楔形切除术显示出比肺叶切除术更好的结果。此外,保存时间少于30天(1个月)的样品比旧样品有更好的成功机会.定义RNA-seq中分析前变量的作用允许检测更适合分析的样品,并更有效地规划NSCLC中基于分子的诊断方法。
    RNA-based next-generation sequencing (RNA-seq) represents the gold standard for detecting gene fusion in non-small cell lung cancer (NSCLC). Despite this, RNA instability makes the management of tissue samples extremely complex, resulting in a significant number of test failures with missing data or the need to switch to other techniques. In the present study, we analyzed pre-analytical variables in 140 tumor tissue samples from patients affected by NSCLC to detect features that increase the chances of successful RNA-seq. We found that the success rate of the analysis positively correlates with the RNA concentration and fragmentation index. Interestingly, small biopsies were more suitable samples than surgical specimens and cell blocks. Among surgical specimens, wedge resections demonstrated better results than lobectomy. Moreover, samples stored for less than 30 days (1 month) had a better chance of success than older samples. Defining the role of pre-analytical variables in RNA-seq allows the detection of more suitable samples for analysis and more effective planning of molecular-based diagnostic approaches in NSCLC.
    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
    脊髓性肌萎缩症(SMA)是一种主要由SMN1基因突变引起的遗传性疾病,导致运动神经元变性和肌肉萎缩,影响多个器官系统。Nusinersen治疗靶向基因表达,有望增强四肢和躯干随意肌的运动功能。运动技能可以通过特定的量表进行评估,例如修订的上肢模块量表(RULM)和扩展的Hammersmith功能运动量表(HFMSE)。这项研究旨在评估nusinersen对SMA2型和3型患者运动技能的影响,使用54个月收集的真实世界数据。对37例接受nusinersen治疗的SMA患者进行了一项前瞻性纵向研究,用R统计软件进行数据分析。结果显示运动功能有显著改善,特别是在具有较高RULM和HFSME评分的SMA3型患者中。此外,GEE分析确定时间,type,年龄,和外显子缺失是运动评分改善的重要预测因子。观察期的延长既是本研究的主要优势,也是局限性,因为辍学率可能会给口译带来挑战。反应的可变性,受遗传背景的影响,SMA类型,和发病年龄,强调了对个性化治疗方法的需求。
    Spinal muscular atrophy (SMA) is a genetic disorder primarily caused by mutations in the SMN1 gene, leading to motor neuron degeneration and muscle atrophy, affecting multiple organ systems. Nusinersen treatment targets gene expression and is expected to enhance the motor function of voluntary muscles in the limbs and trunk. Motor skills can be assessed through specific scales like the Revised Upper Limb Module Scale (RULM) and Hammersmith Functional Motor Scale Expanded (HFMSE). This study aims to evaluate the influence of nusinersen on the motor skills of patients with SMA Type 2 and 3 using real-world data collected over 54 months. A prospective longitudinal study was conducted on 37 SMA patients treated with nusinersen, analyzing data with R statistical software. The outcomes revealed significant improvements in motor functions, particularly in SMA Type 3 patients with higher RULM and HFSME scores. Additionally, GEE analysis identified time, type, age, and exon deletions as essential predictors of motor score improvements. The extended observation period is both a major strength and a limitation of this research, as the dropout rates could present challenges in interpretation. Variability in responses, influenced by genetic background, SMA type, and onset age, highlights the need for personalized treatment approaches.
    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
    胰腺癌是一种高度致命的疾病,经常被诊断为晚期,5年总生存率约为10%。目前的治疗效果有限,强调需要新的治疗选择。本综述旨在确定和总结FGFR(成纤维细胞生长因子受体)抑制剂的临床前和临床研究,包括酪氨酸激酶抑制剂(TKIs)和FGFR特异性抑制剂,在FGFR改变的胰腺癌中。我们纳入了分析疗效的研究,安全,和不同人群的生存结果。对主要数据库的全面搜索确定了73项相关研究:32项临床前研究,16临床,和25来自灰色文学。临床试验主要集中在疗效(20项研究)和安全性(14项研究),解决生存结果的研究较少。FGFR1是研究最多的改变,其次是FGFR2和FGFR4。尽管FGFR改变在胰腺癌中相对罕见,现有数据,包括有希望的现实生活结果,提示FGFR抑制剂的巨大潜力。然而,需要更广泛的研究来确定正确的遗传驱动因素并收集可靠的生存数据。正在进行和未来的试验有望提供更全面的见解,可能导致改善FGFR改变的胰腺癌患者的靶向治疗。
    Pancreatic cancer is a highly lethal disease, often diagnosed at advanced stages, with a 5-year overall survival rate of around 10%. Current treatments have limited effectiveness, underscoring the need for new therapeutic options. This scoping review aims to identify and summarize preclinical and clinical studies on FGFR (Fibroblast Growth Factor Receptor) inhibitors, including tyrosine kinase inhibitors (TKIs) and FGFR-specific inhibitors, in pancreatic cancer with FGFR alterations. We included studies analyzing efficacy, safety, and survival outcomes in various populations. A comprehensive search across major databases identified 73 relevant studies: 32 preclinical, 16 clinical, and 25 from gray literature. The clinical trials focused primarily on efficacy (20 studies) and safety (14 studies), with fewer studies addressing survival outcomes. FGFR1 was the most studied alteration, followed by FGFR2 and FGFR4. Although FGFR alterations are relatively rare in pancreatic cancer, the available data, including promising real-life outcomes, suggest significant potential for FGFR inhibitors. However, more extensive research is needed to identify the correct genetic drivers and gather robust survival data. Ongoing and future trials are expected to provide more comprehensive insights, potentially leading to improved targeted therapies for pancreatic cancer patients with FGFR alterations.
    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
    肺癌,包括非小细胞肺癌和小细胞肺癌,仍然是全球癌症相关死亡的主要原因,占2020年癌症死亡总数的18%。许多患者已经被确定为晚期转移性疾病,并且预后恶化。对肺癌的遗传理解的最新进展为个性化治疗和靶向治疗开辟了新途径。这篇综述探讨了肺癌遗传学的最新发现,讨论关键的生物标志物,并根据这些遗传信息分析当前的临床治疗方法。最后将讨论未来的前景和潜在的研究方向。
    Lung cancer, including both non-small cell lung cancer and small cell lung cancer, remains the leading cause of cancer-related mortality worldwide, representing 18% of the total cancer deaths in 2020. Many patients are identified already at an advanced stage with metastatic disease and have a worsening prognosis. Recent advances in the genetic understanding of lung cancer have opened new avenues for personalized treatments and targeted therapies. This review examines the latest discoveries in the genetics of lung cancer, discusses key biomarkers, and analyzes current clinical therapies based on this genetic information. It will conclude with a discussion of future prospects and potential research directions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号