Tumor therapy

肿瘤治疗
  • 文章类型: Journal Article
    铁凋亡是2012年发现的一种铁依赖性细胞死亡形式。它包括三个基本生物学途径的协调编排:铁稳态,谷胱甘肽调节和脂质代谢。头颈癌(HNC)是一组发生在上呼吸道和消化道粘膜表面的异质性癌症。头颈部鳞状细胞癌是最常见的HNC类型,占HNC病例的90%以上,发病率和死亡率都很高。尽管在诊断和治疗方面有所改善,5年生存率徘徊在令人沮丧的50-60%,复发困扰着近30%的患者,强调目前可用治疗方法的不足之处。值得注意的是,探索铁性与HNC之间联系的研究仍然很少;然而,本综述努力综合当前有关铁中毒的知识。本综述阐述了铁凋亡的正常生理作用,并讨论了其在HNC发病机制中的潜在参与。还回顾了靶向铁性凋亡的HNC的治疗策略和预后范例。这篇综述旨在为催化HNC中铁凋亡的未来研究提供方向。
    Ferroptosis is an iron-dependent form of cell death that was discovered in 2012. It encompasses the coordinated orchestration of three fundamental biological pathways: Iron homeostasis, glutathione regulation and lipid metabolism. Head and neck cancer (HNC) is a heterogeneous group of cancers occurring on the mucosal surfaces of the upper respiratory and digestive tracts. Head and neck squamous cell carcinoma is the most common type of HNC, accounting for >90% of HNC cases, and has high morbidity and mortality rates. Despite improvements in diagnosis and treatment, the 5-year survival rate hovers at a dismal 50-60%, with recurrence afflicting nearly 30% of patients, highlighting the inadequacies of currently available treatments. Of note, research exploring the nexus between ferroptosis and HNC remains scarce; however, the present review endeavors to synthesize current knowledge surrounding ferroptosis. The present review elaborated on the normal physiological role of ferroptosis and discussed its potential involvement in HNC pathogenesis. Therapeutic strategies and prognostic paradigms for HNC that target ferroptosis were also reviewed. This review aims to provide direction to catalyze future investigations into ferroptosis in HNC.
    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
    氨肽酶是催化氨基酸残基从蛋白质或肽底物的N-末端片段裂解的外肽酶。由于其功能,它们在蛋白质成熟中起重要作用,信号转导,细胞周期控制,和各种疾病机制,特别是在癌症病理学中。为了更好地了解它们的功能,荧光和生物/化学发光探针辅助分子成像已成为其不可缺少的优势,包括出色的灵敏度,选择性,以及实时和非侵入性成像。做出了许多努力来开发可有效提高效率和准确性以及最小化副作用的可激活探针。这篇综述是根据氨基肽酶的类型进行分类的,总结了一些最近的设计作品,工作机制,和感应,成像,和他们的可激活探针的治疗性能。最后,概述了目前在开发用于氨肽酶的可激活探针方面的挑战,并为未来的发展提供了可能的解决方案.
    Aminopeptidases are exopeptidases that catalyze the cleavage of amino acid residues from the N-terminal fragment of protein or peptide substrates. Owing to their function, they play important roles in protein maturation, signal transduction, cell-cycle control, and various disease mechanisms, notably in cancer pathology. To gain better insights into their function, molecular imaging assisted by fluorescence and bio/chemiluminescence probes has become an indispensable method to their superiorities, including excellent sensitivity, selectivity, and real-time and noninvasive imaging. Numerous efforts are made to develop activatable probes that can effectively enhance efficiency and accuracy as well as minimize the side effects. This review is classified according to the type of aminopeptidases, summarizing some recent works on the design, work mechanism, and sensing, imaging, and theranostic performance of their activatable probe. Finally, the current challenges are outlined in developing activatable probes for aminopeptidases and provide possible solutions for future advancements.
    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
    免疫抑制性肿瘤微环境和有限的肿瘤内渗透在很大程度上限制了肿瘤治疗的结果。在这里,我们报道了一种定制的基于DNA结构的纳米平台,该平台具有惊人的肿瘤穿透能力,可在体内靶向重塑免疫抑制肿瘤微环境.在我们的设计中,化学免疫调节剂(吉西他滨)可以通过活性氧(ROS)敏感性接头精确地移植到DNA序列中。自组装后,吉西他滨嫁接的DNA结构可以在每个顶点上位点特异性地组织可激活的蜂毒素前体肽(promelittin),用于肿瘤内递送,并进一步充当模板以加载光敏剂(亚甲蓝)用于ROS生产。量身定制的DNA纳米平台可以实现定向积累,高度改善的肿瘤内渗透,和通过激光照射肿瘤细胞的有效免疫原性细胞死亡。最后,通过减少多型免疫抑制细胞和增强细胞毒性淋巴细胞在肿瘤中的浸润,可以成功地重塑免疫抑制肿瘤微环境。这种合理开发的多功能DNA纳米平台为肿瘤治疗的发展提供了新的途径。
    Immunosuppressive tumor microenvironment and limited intratumoral permeation have largely constrained the outcome of tumor therapy. Herein, we report a tailored DNA structure-based nanoplatform with striking tumor-penetrating capability for targeted remodeling of immunosuppressive tumor microenvironment in vivo. In our design, chemo-immunomodulator (gemcitabine) can be precisely grafted in DNA sequences via a reactive oxygen species (ROS)-sensitive linker. After self-assembly, the gemcitabine-grafted DNA structure can site-specifically organize legumain-activatable melittin pro-peptide (promelittin) on each vertex for intratumoral delivery and further function as the template to load photosensitizers (methylene blue) for ROS production. The tailored DNA nanoplatform can achieve targeted accumulation, highly improved intratumoral permeation, and efficient immunogenic cell death of tumor cells by laser irradiation. Finally, the immunosuppressive tumor microenvironment can be successfully remodeled by reducing multi-type immunosuppressive cells and enhancing the infiltration of cytotoxic lymphocytes in the tumor. This rationally developed multifunctional DNA nanoplatform provides a new avenue for the development of tumor therapy.
    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
    代谢重编程为肿瘤提供了能量来源和生物燃料,以支持其在恶性微环境中的生存。对肿瘤微环境(TME)的内在致癌机制的广泛研究已经确定,癌症相关成纤维细胞(CAFs)和代谢重编程通过许多生物学活性调节肿瘤进展。包括肿瘤免疫抑制,慢性炎症,生态位重塑。具体来说,通过共同支持慢性炎症的CAFs和多种免疫细胞促进免疫抑制性TME形成,释放介质,从而诱导转移前生态位的形成,并最终推动肿瘤增殖和转移的恶性循环。本文综述了肿瘤适应性TME动态演化的CAFs和代谢调控过程,特别关注CAFs促进免疫抑制微环境形成和支持转移的机制。现有的发现证实了TME的多个组成部分协同作用以加速肿瘤事件的进展。在推进与CAF相关的研究的背景下,进一步讨论了基于CAF的靶向治疗在临床环境中的潜在应用和挑战。
    Metabolic reprogramming provides tumors with an energy source and biofuel to support their survival in the malignant microenvironment. Extensive research into the intrinsic oncogenic mechanisms of the tumor microenvironment (TME) has established that cancer-associated fibroblast (CAFs) and metabolic reprogramming regulates tumor progression through numerous biological activities, including tumor immunosuppression, chronic inflammation, and ecological niche remodeling. Specifically, immunosuppressive TME formation is promoted and mediators released via CAFs and multiple immune cells that collectively support chronic inflammation, thereby inducing pre-metastatic ecological niche formation, and ultimately driving a vicious cycle of tumor proliferation and metastasis. This review comprehensively explores the process of CAFs and metabolic regulation of the dynamic evolution of tumor-adapted TME, with particular focus on the mechanisms by which CAFs promote the formation of an immunosuppressive microenvironment and support metastasis. Existing findings confirm that multiple components of the TME act cooperatively to accelerate the progression of tumor events. The potential applications and challenges of targeted therapies based on CAFs in the clinical setting are further discussed in the context of advancing research related to CAFs.
    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
    工程生物杂种最近已成为癌症治疗应用的创新仿生平台。特别是,工程光响应生物杂种具有巨大的潜力对抗肿瘤,由于其有趣的仿生特性,光响应能力,和增强的生物治疗功能。在这次审查中,综述了工程光响应生物杂交体的设计原理及其在肿瘤治疗中的最新进展。突出了代表性的工程光响应生物杂种,包括生物分子相关,基于细胞膜,真核细胞为基础,以细菌为基础,和基于藻类的光响应生物杂种。介绍了工程光响应生物杂种的代表性肿瘤治疗方式,包括光热疗法,光动力疗法,协同治疗,和肿瘤治疗结合组织再生。此外,讨论了这些光响应生物杂种在临床实践中的挑战和未来前景。
    Engineered biohybrids have recently emerged as innovative biomimetic platforms for cancer therapeutic applications. Particularly, engineered photoresponsive biohybrids hold tremendous potential against tumors due to their intriguing biomimetic properties, photoresponsive ability, and enhanced biotherapeutic functions. In this review, the design principles of engineered photoresponsive biohybrids and their latest progresses for tumor therapy are summarized. Representative engineered photoresponsive biohybrids are highlighted including biomolecules-associated, cell membrane-based, eukaryotic cell-based, bacteria-based, and algae-based photoresponsive biohybrids. Representative tumor therapeutic modalities of the engineered photoresponsive biohybrids are presented, including photothermal therapy, photodynamic therapy, synergistic therapy, and tumor therapy combined with tissue regeneration. Moreover, the challenges and future perspectives of these photoresponsive biohybrids for clinical practice are discussed.
    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
    肿瘤成像和治疗的主要挑战围绕着提高靶向效率,增强探针/药物递送功效,最小化脱靶信号和毒性。尽管已经开发了各种载体,许多难以合成,昂贵的,并不普遍适用。此外,许多载体在实体瘤中表现出有限的递送率,尤其是较大的纳米载体。为了应对这些挑战,使用容易合成的小分子Cys(SEt)-Lys-CBT(CKCBT)作为自组装构建块,设计了一种简单的二元共组装药物递送平台。CKCBT由于其带正电荷的Lys侧链和较小的尺寸,可以有效地穿透肿瘤细胞。谷胱甘肽还原后,CKCBT与尼罗红或氯蛋白e6共组装以在肿瘤细胞内形成纳米纤维。这使得它们在肿瘤细胞而不是正常细胞中的特异性积累,并延长了它们的暴露时间,导致精确和增强的肿瘤成像和治疗。因此,这种简单高效的二元共组装药物递送平台可以很容易地适应广谱的探针和药物,为推进临床诊断和治疗提供了一种新的方法。
    The primary challenges in tumor imaging and therapy revolve around improving targeting efficiency, enhancing probe/drug delivery efficacy, and minimizing off-target signals and toxicity. Although various carriers have been developed, many are difficult to synthesize, costly, and not universally applicable. Furthermore, numerous carriers exhibit limited delivery rates in solid tumors, particularly larger nanocarriers. To address these challenges, a simple binary co-assembly drug delivery platform has been designed using the readily synthesized small molecule Cys(SEt)-Lys-CBT (CKCBT) as the self-assembly building block. CKCBT can effectively penetrate tumor cells due to its positively charged Lys side chain and small size. Upon glutathione reduction, CKCBT co-assembles with Nile red or Chlorin e6 to form nanofibers inside tumor cells. This enables their specific accumulation in tumor cells rather than normal cells and extends their exposure time, resulting in precise and enhanced tumor imaging and treatment. Hence, this uncomplicated and highly efficient binary co-assembly drug delivery platform can be easily adapted to a broad spectrum of probes and drugs, presenting a novel approach for advancing clinical diagnosis and therapy.
    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
    DNA相分离参与染色质包装以调节基因转录,但是在活细胞中诱导DNA相分离以治疗疾病面临着巨大的挑战。在这里,我们构建了Ru(II)-聚吡啶-负载的上转换纳米平台(表示为UCSNs-R),以实现DNA相分离的操纵和丰富的单线态氧(1O2)的产生,以有效治疗神经胶质瘤。UCSN的利用不仅促进Ru(II)-聚吡啶配合物(RuC)的高负载,而且促进近红外(NIR)激光向紫外光的转化以有效地产生1O2。释放的RuC表现出DNA“光开关”行为和高DNA结合亲和力,诱导活细胞中DNA的相分离,从而导致DNA损伤并抑制肿瘤细胞生长。体内研究表明,在NIR激光照射下,UCSN-R具有很高的抑制肿瘤增殖的能力。这项工作代表了通过将UCSN与Ru(II)-聚吡啶基复合物整合来设计DNA相分离纳米诱导物的范例,以有效治疗神经胶质瘤。
    DNA phase separation participates in chromatin packing for the modulation of gene transcription, but the induction of DNA phase separation in living cells for disease treatment faces huge challenges. Herein, we construct a Ru(II)-polypyridyl-loaded upconversion nanoplatform (denoted as UCSNs-R) to achieve the manipulation of DNA phase separation and production of abundant singlet oxygen (1O2) for efficient treatment of gliomas. The utilization of the UCSN not only facilitates high loading of Ru(II)-polypyridyl complexes (RuC) but also promotes the conversion of near-infrared (NIR) laser to ultraviolet light for efficient 1O2 generation. The released RuC exhibit DNA \"light-switch\" behavior and high DNA binding affinity that induce phase separation of DNA in living cells, thus resulting in DNA damage and suppressing tumor-cell growth. In vivo investigation demonstrates the high capability of UCSNs-R in inhibiting tumor proliferation under NIR laser illumination. This work represents a paradigm for designing a DNA phase separation nanoinducer through integration of the UCSN with Ru(II)-polypyridyl-based complexes for efficient therapy of gliomas.
    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:白血病抑制因子(LIF)是IL-6细胞因子家族的多功能成员,通过与细胞表面上由LIFR和gp130组成的异二聚体结合来激活下游信号传导途径。先前的研究表明,LIF在各种肿瘤组织中高表达(例如胰腺癌,乳腺癌,前列腺癌,和结肠直肠癌)并促进癌细胞增殖,迁移,入侵,和差异化。此外,LIF的过度表达与不良的临床病理特征相关。因此,我们假设LIF可能是一个有希望的癌症治疗靶点.在这项工作中,我们开发了针对LIF的拮抗剂抗体1G11,并在小鼠模型中研究了其抗肿瘤机制及其治疗效果。
    结果:从幼稚的人scFv噬菌体文库中筛选了一系列靶向LIF的单链可变片段(scFv)。这些scFv以完全IgG形式重建并由哺乳动物瞬时表达系统产生。在抗体中,1G11对人表现出优异的结合活性,食蟹猴和小鼠LIF。功能分析表明1G11可以阻断LIF与LIFR的结合并抑制细胞内STAT3磷酸化信号。有趣的是,1G11不阻断LIF与gp130的结合,gp130是另一种参与与LIFR-起形成受体复合物的LIF受体。在体内,在结直肠癌的CT26和MC38模型中,腹膜内施用1G11抑制肿瘤生长。IHC分析显示肿瘤组织中p-STAT3和Ki67降低,而c-caspase3增加。此外,1G11治疗改善CD3+,肿瘤组织中CD4+和CD8+T细胞浸润。
    结论:我们从幼稚的人scFv噬菌体文库中开发了靶向LIF/LIFR信号通路的拮抗剂抗体。拮抗剂抗LIF抗体通过特异性降低p-STAT3发挥抗肿瘤作用。进一步的研究表明,抗LIF抗体1G11增加了肿瘤组织中的免疫细胞浸润。
    BACKGROUND: Leukemia inhibitory factor (LIF) is a multifunctional member of the IL-6 cytokine family that activates downstream signaling pathways by binding to the heterodimer consisting of LIFR and gp130 on the cell surface. Previous research has shown that LIF is highly expressed in various tumor tissues (e.g. pancreatic cancer, breast cancer, prostate cancer, and colorectal cancer) and promotes cancer cell proliferation, migration, invasion, and differentiation. Moreover, the overexpression of LIF correlates with poor clinicopathological characteristics. Therefore, we hypothesized that LIF could be a promising target for the treatment of cancer. In this work, we developed the antagonist antibody 1G11 against LIF and investigated its anti-tumor mechanism and its therapeutic efficacy in mouse models.
    RESULTS: A series of single-chain variable fragments (scFvs) targeting LIF were screened from a naive human scFv phage library. These scFvs were reconstructed in complete IgG form and produced by the mammalian transient expression system. Among the antibodies, 1G11 exhibited the excellent binding activity to human, cynomolgus monkey and mouse LIF. Functional analysis demonstrated 1G11 could block LIF binding to LIFR and inhibit the intracellular STAT3 phosphorylation signal. Interestingly, 1G11 did not block LIF binding to gp130, another LIF receptor that is involved in forming the receptor complex together with LIFR. In vivo, intraperitoneal administration of 1G11 inhibited tumor growth in CT26 and MC38 models of colorectal cancer. IHC analysis demonstrated that p-STAT3 and Ki67 were decreased in tumor tissue, while c-caspase 3 was increased. Furthermore, 1G11 treatment improves CD3+, CD4 + and CD8 + T cell infiltration in tumor tissue.
    CONCLUSIONS: We developed antagonist antibodies targeting LIF/LIFR signaling pathway from a naive human scFv phage library. Antagonist anti-LIF antibody exerts antitumor effects by specifically reducing p-STAT3. Further studies revealed that anti-LIF antibody 1G11 increased immune cell infiltration in tumor tissues.
    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
    角化是一种新发现的由细胞内过量铜(Cu)积累诱导的细胞死亡形式。机械上,铜诱导的二氢硫磺酰胺S-乙酰转移酶聚集的结果,与线粒体三羧酸循环和铁硫簇蛋白的丢失有关,最终导致蛋白毒性应激并触发细胞死亡。最近,由于其作为一种重要的癌症治疗策略的潜力,因此在肿瘤研究中引起了极大的兴趣。在这次审查中,我们总结了细胞凋亡的细胞和分子机制及其与其他类型细胞死亡的关系。此外,我们回顾了目前可用于诱导肿瘤细胞角化的药物或策略,包括铜离子载体,小化合物,和纳米医学。此外,我们在癌症治疗中靶向细胞代谢和特异性调控基因,以增强肿瘤对角化的敏感性。最后,我们讨论了靶向性细胞凋亡克服肿瘤化疗和免疫治疗耐药的可行性,并提出了未来的研究方向。这项研究表明,靶向角化可以为发展肿瘤治疗开辟新的途径。
    Cuproptosis is a newly identified form of cell death induced by excessive copper (Cu) accumulation within cells. Mechanistically, cuproptosis results from Cu-induced aggregation of dihydrolipoamide S-acetyltransferase, correlated with the mitochondrial tricarboxylic acid cycle and the loss of iron-sulfur cluster proteins, ultimately resulting in proteotoxic stress and triggering cell death. Recently, cuproptosis has garnered significant interest in tumor research due to its potential as a crucial therapeutic strategy against cancer. In this review, we summarized the cellular and molecular mechanisms of cuproptosis and its relationship with other types of cell death. Additionally, we reviewed the current drugs or strategies available to induce cuproptosis in tumor cells, including Cu ionophores, small compounds, and nanomedicine. Furthermore, we targeted cell metabolism and specific regulatory genes in cancer therapy to enhance tumor sensitivity to cuproptosis. Finally, we discussed the feasibility of targeting cuproptosis to overcome tumor chemotherapy and immunotherapy resistance and suggested future research directions. This study suggested that targeting cuproptosis could open new avenues for developing tumor 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
    作为一种有前途的基因治疗策略,肿瘤细胞中可控的小分子-mRNA共价修饰可以通过单线态氧(1O2)启动来完成修饰过程。然而,体内1O2的产生通常依赖于外界光的激发,和组织的有限的光穿透极大地干扰了深部肿瘤光疗的发展。这里,我们构建了一个肿瘤靶向纳米胶束,用于细胞内自发生成1O2,而不需要外界光,并在肿瘤细胞中诱导高水平的mRNA共价修饰。Luminal和Ce6通过肿瘤微环境中高水平的过氧化氢(H2O2)触发的化学发光共振能量转移(CRET)化学键合产生1O2。足够的1O2将负载的呋喃氧化成高反应性的二羰基部分,与腺嘌呤(A)进行环加成反应,胞嘧啶(C)或鸟嘌呤(G)对肿瘤细胞蛋白表达的mRNA干扰,从而抑制肿瘤进展。体外和体内实验表明,这种自我启动的基因治疗纳米胶束可以在没有外部光的情况下诱导1O2对mRNA的共价修饰,这个过程可以通过荧光成像实时监控,为基于RNA的肿瘤基因治疗提供了有效的策略。
    As a promising gene therapy strategy, controllable small molecule-mRNA covalent modification in tumor cells could be initiated by singlet oxygen (1O2) to complete the modification process. However, in vivo generation of 1O2 is usually dependent on excitation of external light, and the limited light penetration of tissues greatly interferes the development of deep tumor phototherapy. Here, we constructed a tumor-targeting nano-micelle for the spontaneous intracellular generation of 1O2 without the need for external light, and inducing a high level of covalent modification of mRNA in tumor cells. Luminal and Ce6 were chemically bonded to produce 1O2 by chemiluminescence resonance energy transfer (CRET) triggered by high levels of hydrogen peroxide (H2O2) in the tumor microenvironment. The sufficient 1O2 oxidized the loaded furan to highly reactive dicarbonyl moiety, which underwent cycloaddition reaction with adenine (A), cytosine (C) or guanine (G) on the mRNA for interfering with the tumor cell protein expression, thereby inhibiting tumor progression. In vitro and in vivo experiments demonstrated that this self-initiated gene therapy nano-micelle could induce covalent modification of mRNA by 1O2 without external light, and the process could be monitored in real time by fluorescence imaging, which provided an effective strategy for RNA-based tumor gene therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号