targeted therapeutics

靶向治疗
  • 暂无摘要。
    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
    黑色素瘤是最致命的皮肤癌,每年全世界约有60,000人丧生。尽管有靶向治疗的发展,包括对仅在癌细胞中表达的突变癌蛋白具有选择性的化合物,许多患者要么对初始治疗无反应,要么肿瘤出现耐药性。这导致5年生存率低于25%。杀死耐药黑色素瘤细胞或阻止其出现的新策略将是非常有价值的。黑色素瘤,像其他癌症一样,长期以来一直被描述为处于增加的氧化应激下,导致对抗氧化防御系统的依赖增加。氧化还原稳态的变化在转移期间和与治疗抗性发展相关的代谢重编程期间最为明显。这篇综述讨论了黑素瘤中的氧化应激,特别关注靶向抗氧化途径以限制耐药细胞的出现。
    Melanoma is the deadliest form of skin cancer, with the loss of approximately 60,000 lives world-wide each year. Despite the development of targeted therapeutics, including compounds that have selectivity for mutant oncoproteins expressed only in cancer cells, many patients are either unresponsive to initial therapy or their tumors acquire resistance. This results in five-year survival rates of below 25%. New strategies that either kill drug-resistant melanoma cells or prevent their emergence would be extremely valuable. Melanoma, like other cancers, has long been described as being under increased oxidative stress, resulting in an increased reliance on antioxidant defense systems. Changes in redox homeostasis are most apparent during metastasis and during the metabolic reprogramming associated with the development of treatment resistance. This review discusses oxidative stress in melanoma, with a particular focus on targeting antioxidant pathways to limit the emergence of drug resistant cells.
    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: Congress
    肿瘤学的进步,尤其是在精准肿瘤学时代,导致癌症治疗的范式转变。的确,创新技术,比如人工智能,正在为加强诊断铺平道路,预防,和个性化治疗以及新的药物发现。尽管取得了巨大的进步,耐药癌症的出现限制了我们前进的速度和程度。通过结合他们对基本生物学机制和人工智能和数据科学等技术进步的理解,癌症研究人员现在开始解决这个问题。一起,这将彻底改变癌症治疗,通过加强可能有助于癌症预防的分子干预,告知临床决策,并加速新型治疗药物的开发。这里,我们将讨论人工智能和精准肿瘤学的进展和方法,在第59届爱尔兰癌症研究协会年度会议上发表。
    Advancements in oncology, especially with the era of precision oncology, is resulting in a paradigm shift in cancer care. Indeed, innovative technologies, such as artificial intelligence, are paving the way towards enhanced diagnosis, prevention, and personalised treatments as well as novel drug discoveries. Despite excellent progress, the emergence of resistant cancers has curtailed both the pace and extent to which we can advance. By combining both their understanding of the fundamental biological mechanisms and technological advancements such as artificial intelligence and data science, cancer researchers are now beginning to address this. Together, this will revolutionise cancer care, by enhancing molecular interventions that may aid cancer prevention, inform clinical decision making, and accelerate the development of novel therapeutic drugs. Here, we will discuss the advances and approaches in both artificial intelligence and precision oncology, presented at the 59th Irish Association for Cancer Research annual conference.
    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
    当我们进入精准医学时代,遗传改变与前列腺癌(PCa)发展和治疗的相关性日益增加,这表明在基因组水平上表征临床前模型的重要性.我们的研究调查了8种PCa细胞系的基因组特征,以了解哪些模型是临床相关的。我们设计了一个定制的AmpliSeqDNA基因组,涵盖了靶向AR信号传导的关键分子途径,凋亡,DNA损伤修复,和PI3K/AKT/PTEN,除了肿瘤抑制基因。我们检查了细胞系基因组改变与治疗反应之间的关系。此外,使用DepMap的Celligner工具,我们确定了哪些临床前模型在cBioPortal上最能代表特定前列腺癌患者人群.这些数据将帮助研究人员了解PCa临床前模型的遗传差异,并确定哪些与转化研究相关。
    As we move into the era of precision medicine, the growing relevance of genetic alterations to prostate cancer (PCa) development and treatment demonstrates the importance of characterizing preclinical models at the genomic level. Our study investigated the genomic characterization of eight PCa cell lines to understand which models are clinically relevant. We designed a custom AmpliSeq DNA gene panel that encompassed key molecular pathways targeting AR signaling, apoptosis, DNA damage repair, and PI3K/AKT/PTEN, in addition to tumor suppressor genes. We examined the relationship between cell line genomic alterations and therapeutic response. In addition, using DepMap\'s Celligner tool, we identified which preclinical models are most representative of specific prostate cancer patient populations on cBioPortal. These data will help investigators understand the genetic differences in preclinical models of PCa and determine which ones are relevant for use in their translational research.
    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
    基质金属蛋白酶-1(MMP-1)在牙周病的发病机制中起着举足轻重的作用,特别是牙周炎,凭借其针对I型胶原蛋白的胶原分解活性,牙周组织的主要成分。这篇综述摘要阐明了MMP-1在牙周组织稳态中的复杂参与及其在疾病状态中的失调。MMP-1水平升高,在牙周炎患者的牙龈组织和沟液中观察到,与牙周膜内胶原纤维的降解有关。这种降解有助于牙齿从周围组织脱离并加剧牙槽骨吸收,牙周破裂的标志特征。治疗学上,靶向MMP-1活性成为一种有前途的策略,促使正在进行的研究MMP抑制剂和宿主调节疗法。了解MMP-1在牙周病中的微妙作用为个性化治疗方法铺平了道路,并有望重塑牙周病管理以改善患者预后和牙周健康。
    Matrix metalloproteinase-1 (MMP-1) plays a pivotal role in the pathogenesis of periodontal diseases, particularly periodontitis, by virtue of its collagenolytic activity targeting collagen type I, the primary component of periodontal tissues. This review abstract elucidates the intricate involvement of MMP-1 in periodontal tissue homeostasis and its dysregulation in disease states. Elevated MMP-1 levels, observed in gingival tissues and crevicular fluid of individuals with periodontitis, correlate with the degradation of collagen fibers within the periodontium. This degradation contributes to the detachment of teeth from surrounding tissues and exacerbates alveolar bone resorption, hallmark features of periodontal breakdown. Therapeutically, targeting MMP-1 activity emerges as a promising strategy, prompting ongoing research into MMP inhibitors and host modulation therapies. Understanding MMP-1\'s nuanced role in periodontal diseases paves the way for personalized treatment approaches and holds promise in reshaping periodontal disease management for improved patient outcomes and periodontal health.
    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
    我们介绍了一例成年男性,他患有全血细胞减少症并伴有有症状的贫血,需要慢性输血。他被诊断为系统性肥大细胞增多症并伴有血液肿瘤。在对midostaurin治疗反应不足后,患者开始使用新批准的阿伐替尼.患者在所有三种血细胞系中均显示出显着改善;然而,他出现了腿部水肿,blepheredema,和牙龈出血的药物。该病例强调了治疗晚期系统性肥大细胞增多症患者的复杂性,高选择性KIT抑制在其治疗中的新兴作用,以及药物不良反应的实际管理。
    We present a case of an adult male who presented with pancytopenia accompanied by symptomatic anemia, necessitating chronic transfusions. He was diagnosed with systemic mastocytosis with an associated hematologic neoplasm. Following an inadequate response to midostaurin therapy, the patient was initiated on the newly approved avapritinib. The patient showed significant improvements in all three blood cell lines; however, he developed leg edema, blepharedema, and gum bleeding on this medication. This case underscores the intricacies of managing a patient with advanced systemic mastocytosis, the emerging role of highly selective KIT inhibition in its treatment, and the practical management of adverse medication effects.
    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
    小儿脑肿瘤是儿童癌症相关死亡的主要原因,髓母细胞瘤(MB)是最常见的类型。对这些恶性肿瘤的更好理解导致它们分为四个主要的分子亚组。这种分类不仅有利于临床试验的分层,而且还开发了更有效的疗法。尽管最近取得了进展,诊断为MB的儿童中约有30%出现肿瘤复发.MB中的复发性疾病通常是转移性的并且对当前疗法的反应较差。因此,只有一小部分复发MB患者存活超过1年.由于其令人沮丧的预后,我们迫切需要以预防或控制疾病复发为目标的新型治疗策略.在这次审查中,我们总结了我们对MB治疗失败背后的分子机制的理解的最新进展,以及那些具有复发性病例特征的病例。我们还提出了如何使用这些发现来更好地为新诊断和复发性MB的个性化医疗方法提供信息的途径。最后,我们讨论目前正在评估的MB患者的治疗方法,特别强调在诊断和复发时按亚组靶向MB的那些。
    Pediatric brain tumors are the leading cause of cancer-related deaths in children, with medulloblastoma (MB) being the most common type. A better understanding of these malignancies has led to their classification into four major molecular subgroups. This classification not only facilitates the stratification of clinical trials, but also the development of more effective therapies. Despite recent progress, approximately 30% of children diagnosed with MB experience tumor relapse. Recurrent disease in MB is often metastatic and responds poorly to current therapies. As a result, only a small subset of patients with recurrent MB survive beyond one year. Due to its dismal prognosis, novel therapeutic strategies aimed at preventing or managing recurrent disease are urgently needed. In this review, we summarize recent advances in our understanding of the molecular mechanisms behind treatment failure in MB, as well as those characterizing recurrent cases. We also propose avenues for how these findings can be used to better inform personalized medicine approaches for the treatment of newly diagnosed and recurrent MB. Lastly, we discuss the treatments currently being evaluated for MB patients, with special emphasis on those targeting MB by subgroup at diagnosis and relapse.
    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
    尽管取得了巨大的成功,靶向治疗如激酶抑制剂(KIs)仍然造成不良事件如心脏毒性。不同KIs引起的心脏毒性的类型和强度存在很大差异,目前的临床前模型不足以预测它。因此,需要开发更简单和快速的模型,用于在临床前阶段筛选新的KIs.因此,我们旨在建立一个快速和强大的临床前动物模型来预测KIs的心脏毒性,并确定一组FDA批准的KIs的比较心脏毒性谱。在用被批准用于治疗各种癌症的10个KIs治疗后,定期进行水蚤的心率测量和存活分析。水蚤的心率以及存活率在KIs之间以剂量和时间依赖性方式变化,表明各种KIs的不同心脏毒性特征。Further,心脏毒性和生存率之间的相关性在10个KIs中也不同。重要的是,索拉非尼和维罗非尼显示最大和最小的心脏毒性,分别。比较的心脏毒性谱也与先前的研究一致,该研究表明水蚤作为一种有价值且相关的动物模型在临床前阶段快速预测新型KIs的心脏毒性。
    Despite significant success, targeted therapeutics such as kinase inhibitors (KIs) still pose adverse events such as the cardiotoxicity. There is a lot of variation in the type and intensity of cardiotoxicity caused by different KIs and current pre-clinical models are inadequate to predict it. Thus, there is a need to develop more simple and rapid models for screening of novel KIs at the pre-clinical step itself. We thus aimed to establish a rapid and robust pre-clinical animal model for predicting cardiotoxicity of KIs and identify comparative cardiotoxicity profiles of a panel of FDA-approved KIs. Heart rate measurement and survival analysis of Daphnia was performed at regular intervals following treatment with ten KIs that were approved for the treatment of various cancers. The heart rates of Daphnia as well as the survival varied between KIs in a dose and time dependent manner suggesting differential cardiotoxicity profiles of various KIs. Further, the correlation between the cardiotoxicity and survival also varied among the ten KIs. Importantly, sorafenib and vemurafenib displayed maximum and least cardiotoxicity, respectively. The comparative cardiotoxicity profiles also are in conformity with the previous studies indicating the utility of Daphnia as a valuable and relevant animal model to rapidly predict the cardiotoxicity of novel KIs at a pre-clinical stage.
    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: Preprint
    上皮性卵巢癌(EOC)可以是高度致命的,非同源重组缺陷(HRD)疾病患者的治疗选择有限。叶酸受体α(FOLR1/FRα)靶向药物已显示出单独或与现有疗法联合使用的希望。但FRα与其他治疗驱动生物标志物的关系尚不清楚。对癌症基因组图谱(TCGA)进行了查询,以评估患有差异表达FRα蛋白的卵巢肿瘤患者的蛋白和mRNA表达和突变负荷,以及针对目前通过FoundationOneCompanionDiagnostics测试的标准324突变的结果,以鉴定感兴趣的靶标。在TCGA中的585个样本中,确定了121例浆液性卵巢肿瘤患者的FRα蛋白表达定量。FRα蛋白表达与FOLR1mRNA表达显著相关(p=7.19×10-14)。高FRα组(Q1)的无进展生存期(PFS)为20.7个月,与低FRα组的16.6个月相比(Q4,Logrank,p=0.886)。总生存期(OS)分别为54.1个月和36.3个月,分别;然而,这一结果并不显著(Q1与Q4,Logrank,p=0.200)。在高表达FRα的肿瘤患者中更常见的突变包括PIK3CA和FGF家族蛋白。FRa靶向剂与PI3K的组合,mTOR,FGF(R)和VEGF抑制剂值得研究以评估其治疗潜力。
    上皮性卵巢癌可以是高度致命的,对于没有BRCA突变或非同源重组缺陷疾病的患者,治疗选择有限。叶酸受体α(FRα)靶向剂在铂敏感和铂耐药卵巢癌的背景下显示出希望,无论是单独还是与现有疗法相结合,但FRα与其他治疗驱动生物标志物的关系尚不清楚。这项研究确定了表达FRα的肿瘤中潜在的可靶向突变,包括PIK3CA和FGF/R家族蛋白,并为FRa靶向剂与PIK3CA的新型组合的未来研究提供了基础,mTOR,FGF/R,和VEGF抑制剂。
    Epithelial ovarian cancer (EOC) can be highly lethal, with limited therapeutic options for patients with non-homologous recombination deficient (HRD) disease. Folate receptor alpha (FOLR1/FRα)-targeting agents have shown promise both alone and in combination with available therapies, but the relationship of FRα to other treatment-driving biomarkers is unknown. The Cancer Genome Atlas (TCGA) was queried to assess protein and mRNA expression and mutational burden in patients with differential FRα protein-expressing ovarian tumors, and the results referenced against the standard 324 mutations currently tested through FoundationOne Companion Diagnostics to identify targets of interest. Of 585 samples within TCGA, 121 patients with serous ovarian tumors for whom FRα protein expression was quantified were identified. FRα protein expression significantly correlated with FOLR1 mRNA expression (p=7.19×1014). Progression free survival (PFS) for the FRα-high group (Q1) was 20.7 months, compared to 16.6 months for the FRα-low group (Q4, Logrank, p=0.886). Overall survival (OS) was 54.1 months versus 36.3 months, respectively; however, this result was not significant (Q1 vs. Q4, Logrank, p=0.200). Mutations more commonly encountered in patients with high FRα-expressing tumors included PIK3CA and FGF family proteins. Combinations of FRα-targeting agents with PI3K, mTOR, FGF(R) and VEGF inhibitors warrant investigation to evaluate their therapeutic potential.
    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
    转移相关蛋白1/蛋白激酶B(MTA1/AKT)信号通路已被证明在促进前列腺肿瘤生长方面具有协同作用。植物多酚的针对性拦截策略,特别是二苯乙烯,对MTA1介导的前列腺癌进展显示出巨大的希望。在这项研究中,我们在磷酸酶和张力蛋白同源物(Pten)缺失(R26MTA1;Ptenf/f)和PC3M前列腺癌细胞的背景下,采用了MTA1过表达的前列腺特异性转基因小鼠模型,该模型概括了晚期前列腺癌的分子通路改变.机械上,在培养的PC3M细胞中,gnetinC(二聚体白藜芦醇)对MTA1的敲除或药理学抑制导致哺乳动物雷帕霉素靶蛋白(mTOR)信号显着失活。在体内,小鼠耐受每天腹膜内治疗gnetinC(7mg/kgbw)12周,没有任何毒性迹象。在患有晚期前列腺癌的小鼠中,用gnetinC处理显著降低细胞增殖和血管生成并促进细胞凋亡。Further,除了降低前列腺上皮细胞中的MTA1水平,gnetinC显着降低前列腺组织中的mTOR信号活性,包括mTOR靶蛋白的活性:p70核糖体蛋白S6激酶(S6K)和真核翻译起始因子4E(elF4E)结合蛋白1(4EBP1)。总的来说,这些发现确立了gnetinC作为一种新的天然化合物,对MTA1/AKT/mTOR激活的前列腺癌具有抗癌特性,具有作为单一疗法的潜力,并可能作为未来临床批准的mTOR途径抑制剂的辅助药物。
    The metastasis-associated protein 1/protein kinase B (MTA1/AKT) signaling pathway has been shown to cooperate in promoting prostate tumor growth. Targeted interception strategies by plant-based polyphenols, specifically stilbenes, have shown great promise against MTA1-mediated prostate cancer progression. In this study, we employed a prostate-specific transgenic mouse model with MTA1 overexpression on the background of phosphatase and tensin homolog (Pten) null (R26MTA1; Ptenf/f) and PC3M prostate cancer cells which recapitulate altered molecular pathways in advanced prostate cancer. Mechanistically, the MTA1 knockdown or pharmacological inhibition of MTA1 by gnetin C (dimer resveratrol) in cultured PC3M cells resulted in the marked inactivation of mammalian target of rapamycin (mTOR) signaling. In vivo, mice tolerated a daily intraperitoneal treatment of gnetin C (7 mg/kg bw) for 12 weeks without any sign of toxicity. Treatment with gnetin C markedly reduced cell proliferation and angiogenesis and promoted apoptosis in mice with advanced prostate cancer. Further, in addition to decreasing MTA1 levels in prostate epithelial cells, gnetin C significantly reduced mTOR signaling activity in prostate tissues, including the activity of mTOR-target proteins: p70 ribosomal protein S6 kinase (S6K) and eukaryotic translational initiation factor 4E (elF4E)-binding protein 1 (4EBP1). Collectively, these findings established gnetin C as a new natural compound with anticancer properties against MTA1/AKT/mTOR-activated prostate cancer, with potential as monotherapy and as a possible adjunct to clinically approved mTOR pathway inhibitors in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号