Molecular Targeted Therapy

分子靶向治疗
  • 文章类型: Journal Article
    T-BOX因子属于进化保守的转录因子家族。T-BOX因子不仅在生长发育中起关键作用,而且还参与免疫。癌症开始,和进步。此外,相同的T-BOX分子在各种发育过程和肿瘤微环境中表现出不同甚至相反的作用。了解环境依赖性T-BOX因子在恶性肿瘤中的多种作用对于揭示T-BOX靶向癌症治疗的潜力至关重要。我们总结了T-BOX因子在不同发育过程中的生理作用及其表达失调时观察到的病理作用。我们还讨论了它们在肿瘤免疫微环境(TIME)中的调节作用以及尚未解决的新出现的问题。本综述将有助于系统全面地了解T-BOX转录因子家族在肿瘤生理学中的重要作用。病理学,和豁免权。目的是提供有价值的信息来支持T-BOX靶向治疗的发展。
    T-BOX factors belong to an evolutionarily conserved family of transcription factors. T-BOX factors not only play key roles in growth and development but are also involved in immunity, cancer initiation, and progression. Moreover, the same T-BOX molecule exhibits different or even opposite effects in various developmental processes and tumor microenvironments. Understanding the multiple roles of context-dependent T-BOX factors in malignancies is vital for uncovering the potential of T-BOX-targeted cancer therapy. We summarize the physiological roles of T-BOX factors in different developmental processes and their pathological roles observed when their expression is dysregulated. We also discuss their regulatory roles in tumor immune microenvironment (TIME) and the newly arising questions that remain unresolved. This review will help in systematically and comprehensively understanding the vital role of the T-BOX transcription factor family in tumor physiology, pathology, and immunity. The intention is to provide valuable information to support the development of T-BOX-targeted therapy.
    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)是世界范围内高度流行的恶性肿瘤,具有复杂的发病机制和治疗挑战。研究表明,甲基转移酶样3(METTL3)通过其靶RNA的甲基化而广泛参与多种肿瘤的发病机制。及其在BC中的作用和机制也被广泛研究。在这次审查中,我们旨在对现有研究进行全面解释,并阐明METTL3与BC之间的关系.这篇综述表明,高水平的METTL3与发病机制有关。预后不良,和BC的耐药性,提示METTL3是潜在的诊断或预后生物标志物和治疗靶点。总的来说,这篇综述提供了对METTL3如何通过RNA甲基化发挥作用的全面理解,为今后的基础研究和临床应用提供了有价值的参考。
    Breast cancer (BC) is a highly prevalent malignancy worldwide, with complex pathogenesis and treatment challenges. Research reveals that methyltransferase-like 3 (METTL3) is widely involved in the pathogenesis of several tumors through methylation of its target RNAs, and its role and mechanisms in BC are also extensively studied. In this review, we aim to provide a comprehensive interpretation of available studies and elucidate the relationship between METTL3 and BC. This review suggests that high levels of METTL3 are associated with the pathogenesis, poor prognosis, and drug resistance of BC, suggesting METTL3 as a potential diagnostic or prognostic biomarker and therapeutic target. Collectively, this review provides a comprehensive understanding of how METTL3 functions through RNA methylation, which provides a valuable reference for future fundamental studies and clinical applications.
    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
    多发性骨髓瘤(MM)是第二常见的浆细胞恶性血液病,以溶骨性骨病变为特征,贫血,高钙血症,肾功能衰竭,和恶性浆细胞的积累。MM的发病机制涉及MM细胞通过可溶性细胞因子和细胞粘附分子与骨髓微环境相互作用,激活各种信号通路,如PI3K/AKT/mTOR,RAS/MAPK,JAK/STAT,Wnt/β-catenin,和NF-κB通路。这些途径的异常激活有助于增殖,生存,迁移,和骨髓瘤细胞的耐药性,使它们成为有吸引力的治疗干预目标。目前,在MM中针对这些信号通路的批准药物是有限的,许多抑制剂和诱导剂仍处于临床前或临床研究阶段。MM的治疗选择包括非靶向药物,如烷化剂,皮质类固醇,免疫调节药物,蛋白酶体抑制剂,和组蛋白脱乙酰酶抑制剂。此外,靶向药物如单克隆抗体,嵌合抗原受体T细胞,双特异性T细胞衔接者,和双特异性抗体正在用于MM治疗。尽管在MM治疗方面取得了重大进展,这种疾病仍然无法治愈,强调需要根据新兴的理论知识开发新的或联合的靶向疗法,技术,和平台。在这次审查中,我们强调了信号通路在MM的恶性进展和治疗中的关键作用,探索靶向治疗和潜在治疗的进展,为改善MM管理和结局提供进一步的见解。
    Multiple myeloma (MM) is the second most common hematological malignancy of plasma cells, characterized by osteolytic bone lesions, anemia, hypercalcemia, renal failure, and the accumulation of malignant plasma cells. The pathogenesis of MM involves the interaction between MM cells and the bone marrow microenvironment through soluble cytokines and cell adhesion molecules, which activate various signaling pathways such as PI3K/AKT/mTOR, RAS/MAPK, JAK/STAT, Wnt/β-catenin, and NF-κB pathways. Aberrant activation of these pathways contributes to the proliferation, survival, migration, and drug resistance of myeloma cells, making them attractive targets for therapeutic intervention. Currently, approved drugs targeting these signaling pathways in MM are limited, with many inhibitors and inducers still in preclinical or clinical research stages. Therapeutic options for MM include non-targeted drugs like alkylating agents, corticosteroids, immunomodulatory drugs, proteasome inhibitors, and histone deacetylase inhibitors. Additionally, targeted drugs such as monoclonal antibodies, chimeric antigen receptor T cells, bispecific T-cell engagers, and bispecific antibodies are being used in MM treatment. Despite significant advancements in MM treatment, the disease remains incurable, emphasizing the need for the development of novel or combined targeted therapies based on emerging theoretical knowledge, technologies, and platforms. In this review, we highlight the key role of signaling pathways in the malignant progression and treatment of MM, exploring advances in targeted therapy and potential treatments to offer further insights for improving MM management and outcomes.
    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: Editorial
    分子医学在理解疾病机制和发现目标干预措施方面开辟了新的视野。更广泛的DNA和RNA测序,免疫组织化学分析,蛋白质组学,和其他分子测试改变了医生管理疾病的方式。癌症基因组图谱计划提出的胃癌分子分类将胃腺癌分为四种亚型。然而,批准临床使用的可用靶标和/或免疫疗法似乎与这些分子亚型分离。直到对分子分类提供的大量数据进行更可靠的解释,在分子治疗时代,临床指南将依靠可用的可操作靶点和已批准的治疗方法来指导临床医生进行癌症治疗.
    Molecular medicine opened new horizons in understanding disease mechanisms and discovering target interventions. The wider availability of DNA and RNA sequencing, immunohistochemical analysis, proteomics, and other molecular tests changed how physicians manage diseases. The gastric cancer molecular classification proposed by The Cancer Genome Atlas Program divides gastric adenocarcinomas into four subtypes. However, the available targets and/or immunotherapies approved for clinical use seem to be dissociated from these molecular subtypes. Until a more reliable interpretation of the stupendous amount of data provided by the molecular classifications is presented, the clinical guidelines will rely on available actionable targets and approved therapies to guide clinicians in conducting cancer management in the era of molecular therapies.
    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
    糖尿病视网膜病变(DR)是糖尿病引起的眼部常见并发症,随着个体年龄的增长,主要与高血糖水平和高血压有关。DR是I型和II型糖尿病的严重微血管并发症,也是视力障碍的主要原因。对抗和阻止DR进展的关键方法在于有效管理糖尿病患者的血糖和血压水平;然而,这很少实现。人类和动物研究都揭示了这种疾病的复杂性质,涉及各种细胞类型和分子。除了光凝,靶向视网膜中VEGF分子以预防异常血管生长的唯一疗法是玻璃体内抗VEGF疗法.然而,很大一部分案件,大约30-40%,对这种治疗没有反应。这篇综述探讨了DR的独特病理生理现象以及可识别的细胞类型和分子,这些细胞类型和分子可用于缓解糖尿病引起的视网膜慢性变化。解决这一领域的重大研究差距对于扩大有效管理DR的治疗选择至关重要。
    Diabetic retinopathy (DR) stands as a prevalent complication in the eye resulting from diabetes mellitus, predominantly associated with high blood sugar levels and hypertension as individuals age. DR is a severe microvascular complication of both type I and type II diabetes mellitus and the leading cause of vision impairment. The critical approach to combatting and halting the advancement of DR lies in effectively managing blood glucose and blood pressure levels in diabetic patients; however, this is seldom achieved. Both human and animal studies have revealed the intricate nature of this condition involving various cell types and molecules. Aside from photocoagulation, the sole therapy targeting VEGF molecules in the retina to prevent abnormal blood vessel growth is intravitreal anti-VEGF therapy. However, a substantial portion of cases, approximately 30-40%, do not respond to this treatment. This review explores distinctive pathophysiological phenomena of DR and identifiable cell types and molecules that could be targeted to mitigate the chronic changes occurring in the retina due to diabetes mellitus. Addressing the significant research gap in this domain is imperative to broaden the treatment options available for managing DR effectively.
    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
    滋养层细胞表面抗原2(Trop2)在一系列实体瘤和多个致癌信号通路的参与者中过度表达,使其成为有吸引力的治疗靶点。在过去的十年里,各种Trop2靶向疗法的快速发展,特别是以抗体-药物偶联物(ADC)的出现为标志,彻底改变了治疗意见有限的Trop2阳性肿瘤患者的结果,例如三阴性乳腺癌(TNBC)。这篇综述全面总结了Trop2靶向治疗的进展,包括ADC,抗体,多特异性药物,免疫疗法,癌症疫苗,和小分子抑制剂,随着对他们设计的深入讨论,行动机制(MOA),和限制。此外,我们强调了这些新兴的Trop2靶向药物的临床研究进展,重点关注其临床应用和对肿瘤的治疗效果。此外,我们提出了未来研究的方向,例如加强我们对Trop2的结构和生物学的理解,探索最佳组合策略,并根据Trop2测试方法定制精确处理。
    Trophoblast cell surface antigen 2 (Trop2) is overexpressed in a range of solid tumors and participants in multiple oncogenic signaling pathways, making it an attractive therapeutic target. In the past decade, the rapid development of various Trop2-targeted therapies, notably marked by the advent of the antibody-drug conjugate (ADC), revolutionized the outcome for patients facing Trop2-positive tumors with limited treatment opinions, such as triple-negative breast cancer (TNBC). This review provides a comprehensive summary of advances in Trop2-targeted therapies, including ADCs, antibodies, multispecific agents, immunotherapy, cancer vaccines, and small molecular inhibitors, along with in-depth discussions on their designs, mechanisms of action (MOAs), and limitations. Additionally, we emphasize the clinical research progress of these emerging Trop2-targeted agents, focusing on their clinical application and therapeutic efficacy against tumors. Furthermore, we propose directions for future research, such as enhancing our understanding of Trop2\'s structure and biology, exploring the best combination strategies, and tailoring precision treatment based on Trop2 testing methodologies.
    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
    原理:代谢的协同重编程主导神经母细胞瘤(NB)的进展。基于阐明代谢重编程的分子机制,开发具有分层指导的NB治疗选择的个性化风险预测方法具有重要的临床意义。方法:采用基于机器学习的多步骤程序,在单细胞和代谢物通量维度阐明了代谢重编程驱动NB恶性进展的协同机制.随后,我们开发了一种有前景的代谢重编程相关预后特征(MPS)和基于MPS分层的个体化治疗方法,并使用临床前模型进行了进一步独立验证.结果:MPS鉴定的MPS-INB显示出明显高于MPS-II对应物的代谢重编程活性。与目前的临床特征相比,MPS显示出更高的准确性[AUC:0.915vs.0.657(MYCN),0.713(INSS阶段),和0.808(INRG分层)]预测预后。AZD7762和依托泊苷被确定为针对MPS-I和IINB的有效治疗剂,分别。随后的生物学测试表明AZD7762基本上抑制了生长,迁移,MPS-INB细胞的侵袭,比MPS-II细胞更有效。相反,依托泊苷对MPS-Ⅱ型NB细胞有较好的治疗作用。更令人鼓舞的是,AZD7762和依托泊苷显著抑制体内皮下肿瘤发生,扩散,MPS-I和MPS-II样本中的肺转移,分别;从而延长荷瘤小鼠的生存期。机械上,AZD7762和依托泊苷诱导的MPS-I和MPS-II细胞凋亡,分别,通过线粒体依赖性途径;MPS-INB通过谷氨酸代谢成瘾和乙酰辅酶A抵抗依托泊苷诱导的细胞凋亡。MPS-INB进展受多种代谢重编程驱动因素的推动,包括多药耐药,免疫抑制和促进肿瘤的炎症微环境。免疫学,MPS-INB通过MIF和THBS信号通路抑制免疫细胞。代谢,MPS-INB细胞的恶性增殖得到了重新编程的谷氨酸代谢的显著支持,三羧酸循环,尿素循环,等。此外,MPS-INB细胞表现出独特的肿瘤促进发育谱系和自我沟通模式,通过发育和自我通讯激活的致癌信号通路增强证明了这一点。结论:本研究为代谢重编程介导的NB恶性进展的分子机制提供了深刻的见解。它还揭示了在新的精确风险预测方法的指导下开发靶向药物,这可能有助于明显改善NB的治疗策略。
    Rationale: Synergic reprogramming of metabolic dominates neuroblastoma (NB) progression. It is of great clinical implications to develop an individualized risk prognostication approach with stratification-guided therapeutic options for NB based on elucidating molecular mechanisms of metabolic reprogramming. Methods: With a machine learning-based multi-step program, the synergic mechanisms of metabolic reprogramming-driven malignant progression of NB were elucidated at single-cell and metabolite flux dimensions. Subsequently, a promising metabolic reprogramming-associated prognostic signature (MPS) and individualized therapeutic approaches based on MPS-stratification were developed and further validated independently using pre-clinical models. Results: MPS-identified MPS-I NB showed significantly higher activity of metabolic reprogramming than MPS-II counterparts. MPS demonstrated improved accuracy compared to current clinical characteristics [AUC: 0.915 vs. 0.657 (MYCN), 0.713 (INSS-stage), and 0.808 (INRG-stratification)] in predicting prognosis. AZD7762 and etoposide were identified as potent therapeutics against MPS-I and II NB, respectively. Subsequent biological tests revealed AZD7762 substantially inhibited growth, migration, and invasion of MPS-I NB cells, more effectively than that of MPS-II cells. Conversely, etoposide had better therapeutic effects on MPS-II NB cells. More encouragingly, AZD7762 and etoposide significantly inhibited in-vivo subcutaneous tumorigenesis, proliferation, and pulmonary metastasis in MPS-I and MPS-II samples, respectively; thereby prolonging survival of tumor-bearing mice. Mechanistically, AZD7762 and etoposide-induced apoptosis of the MPS-I and MPS-II cells, respectively, through mitochondria-dependent pathways; and MPS-I NB resisted etoposide-induced apoptosis by addiction of glutamate metabolism and acetyl coenzyme A. MPS-I NB progression was fueled by multiple metabolic reprogramming-driven factors including multidrug resistance, immunosuppressive and tumor-promoting inflammatory microenvironments. Immunologically, MPS-I NB suppressed immune cells via MIF and THBS signaling pathways. Metabolically, the malignant proliferation of MPS-I NB cells was remarkably supported by reprogrammed glutamate metabolism, tricarboxylic acid cycle, urea cycle, etc. Furthermore, MPS-I NB cells manifested a distinct tumor-promoting developmental lineage and self-communication patterns, as evidenced by enhanced oncogenic signaling pathways activated with development and self-communications. Conclusions: This study provides deep insights into the molecular mechanisms underlying metabolic reprogramming-mediated malignant progression of NB. It also sheds light on developing targeted medications guided by the novel precise risk prognostication approaches, which could contribute to a significantly improved therapeutic strategy for NB.
    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
    炎性肌纤维母细胞瘤(IMT)是一种罕见的肿瘤,具有中度恶性肿瘤,其特征是倾向于复发,但转移率低。诊断挑战来自不同的病理表现,可变症状学,缺乏不同的成像特征。然而,通过间变性淋巴瘤激酶(ALK)基因的融合来鉴定IMT,这在大约70%的病例中存在,与各种聚变伙伴,包括ran结合蛋白2(RANBP2),这可以确认诊断。虽然手术是局部肿瘤的首选方法,晚期或转移性疾病的最佳长期治疗方法很难确定.靶向治疗对于在IMT遗传改变的背景下实现对治疗的持续反应至关重要。克唑替尼,ALK酪氨酸激酶抑制剂(TKI),于2020年获得美国食品和药物管理局(FDA)的正式批准,用于ALK重排治疗IMT。然而,大多数患者面临阻力和疾病进展,需要考虑序贯治疗。放疗与靶向治疗相结合似乎对该适应症有益。免疫疗法也取得了早期有希望的结果,表明联合治疗方法的潜力。然而,仍然缺乏明确的建议。这篇综述分析了关于IMT的现有研究,包括遗传疾病及其对疾病进程的影响,有关最新靶向治疗方案的数据以及在该适应症中开发免疫疗法的可能性,以及总结有关预后和预测因素的一般知识,在对全身治疗的抵抗方面也是如此。
    Inflammatory myofibroblastic tumor (IMT) is a rare neoplasm with intermediate malignancy characterized by a propensity for recurrence but a low metastatic rate. Diagnostic challenges arise from the diverse pathological presentation, variable symptomatology, and lack of different imaging features. However, IMT is identified by the fusion of the anaplastic lymphoma kinase (ALK) gene, which is present in approximately 70% of cases, with various fusion partners, including ran-binding protein 2 (RANBP2), which allows confirmation of the diagnosis. While surgery is the preferred approach for localized tumors, the optimal long-term treatment for advanced or metastatic disease is difficult to define. Targeted therapies are crucial for achieving sustained response to treatment within the context of genetic alteration in IMT. Crizotinib, an ALK tyrosine kinase inhibitor (TKI), was officially approved by the US Food and Drug Administration (FDA) in 2020 to treat IMT with ALK rearrangement. However, most patients face resistance and disease progression, requiring consideration of sequential treatments. Combining radiotherapy with targeted therapy appears to be beneficial in this indication. Early promising results have also been achieved with immunotherapy, indicating potential for combined therapy approaches. However, defined recommendations are still lacking. This review analyzes the available research on IMT, including genetic disorders and their impact on the course of the disease, data on the latest targeted therapy regimens and the possibility of developing immunotherapy in this indication, as well as summarizing general knowledge about prognostic and predictive factors, also in terms of resistance to systemic therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Published Erratum
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号