tumor resistance

肿瘤耐药
  • 文章类型: Journal Article
    光动力疗法(PDT)是一种开创性的方法,涉及在分子氧存在下通过可见光激活光敏剂(PS)在肿瘤内诱导细胞毒性活性氧(ROS)。这种创新疗法已证明在治疗各种癌症方面取得了成功。虽然PDT在大多数实体瘤中非常有效,有迹象表明某些癌症表现出耐药性,一些最初有反应性的癌症可能对PDT产生内在或获得性抗性。这种抗性的分子机制尚不完全清楚。最近的证据表明,类似于其他传统的癌症治疗方法,生存途径的激活,如KEAP1/Nrf2信号通路,正在成为许多癌症中PDT后耐药的重要机制。本文探讨了Nrf2的双重作用,强调了将Nrf2异常表达与一系列癌症的治疗抗性联系起来的证据。此外,它深入研究了Nrf2在癌症光动力疗法中的具体作用,强调Nrf2介导的抗氧化反应上调和药物外排转运体诱导的证据是不同类型癌症抗PDT的潜在机制.因此,了解Nrf2在PDT耐药中的具体作用可能为开发使用PDT的更有效的癌症治疗方法铺平道路.
    Photodynamic therapy (PDT) is a groundbreaking approach involving the induction of cytotoxic reactive oxygen species (ROS) within tumors through visible light activation of photosensitizers (PS) in the presence of molecular oxygen. This innovative therapy has demonstrated success in treating various cancers. While PDT proves highly effective in most solid tumors, there are indications that certain cancers exhibit resistance, and some initially responsive cancers may develop intrinsic or acquired resistance to PDT. The molecular mechanisms underlying this resistance are not fully understood. Recent evidence suggests that, akin to other traditional cancer treatments, the activation of survival pathways, such as the KEAP1/Nrf2 signaling pathway, is emerging as an important mechanism of post-PDT resistance in many cancers. This article explores the dual role of Nrf2, highlighting evidence linking aberrant Nrf2 expression to treatment resistance across a range of cancers. Additionally, it delves into the specific role of Nrf2 in the context of photodynamic therapy for cancers, emphasizing evidence that suggests Nrf2-mediated upregulation of antioxidant responses and induction of drug efflux transporters are potential mechanisms of resistance to PDT in diverse cancer types. Therefore, understanding the specific role(s) of Nrf2 in PDT resistance may pave the way for the development of more effective cancer treatments using PDT.
    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
    肿瘤耐药对有效的癌症治疗提出了重大挑战,探索新的治疗策略势在必行。最近的研究强调了免疫细胞在肿瘤耐药性发展中的重要作用。在肿瘤微环境中,巨噬细胞经历极化为M2表型,从而促进耐药肿瘤的出现。嗜中性粒细胞通过形成细胞外陷阱而有助于肿瘤抗性。而T细胞和自然杀伤(NK)细胞通过对肿瘤细胞的直接细胞毒性发挥其影响。此外,树突状细胞(DC)通过刺激T细胞活化而参与预防肿瘤药物抗性。在这次审查中,我们在分子水平上提供了关于免疫细胞介导的肿瘤抗性调节的现有知识的全面总结,特别关注巨噬细胞,中性粒细胞,DCs,T细胞,NK细胞免疫细胞调节的靶向显示出解决耐药性的巨大潜力,深入了解免疫细胞和肿瘤细胞之间的分子相互作用为创新疗法的发展带来了希望。此外,我们探讨了这些免疫细胞在耐药肿瘤治疗中的临床意义。这篇综述强调了利用免疫细胞的功能能力来有效克服耐药肿瘤的新方法的探索。
    Tumor resistance poses a significant challenge to effective cancer treatment, making it imperative to explore new therapeutic strategies. Recent studies have highlighted the profound involvement of immune cells in the development of tumor resistance. Within the tumor microenvironment, macrophages undergo polarization into the M2 phenotype, thus promoting the emergence of drug-resistant tumors. Neutrophils contribute to tumor resistance by forming extracellular traps. While T cells and natural killer (NK) cells exert their impact through direct cytotoxicity against tumor cells. Additionally, dendritic cells (DCs) have been implicated in preventing tumor drug resistance by stimulating T cell activation. In this review, we provide a comprehensive summary of the current knowledge regarding immune cell-mediated modulation of tumor resistance at the molecular level, with a particular focus on macrophages, neutrophils, DCs, T cells, and NK cells. The targeting of immune cell modulation exhibits considerable potential for addressing drug resistance, and an in-depth understanding of the molecular interactions between immune cells and tumor cells holds promise for the development of innovative therapies. Furthermore, we explore the clinical implications of these immune cells in the treatment of drug-resistant tumors. This review emphasizes the exploration of novel approaches that harness the functional capabilities of immune cells to effectively overcome drug-resistant tumors.
    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
    尽管靶向治疗取得了进展,原发性和获得性耐药性使结直肠癌(CRC)的治疗成为亟待解决的问题。据报道,CRC的发展与miRNA失调有关。多项研究已经证明miR-135b-5p在CRC组织和邻近组织之间具有异常表达水平。然而,目前尚不清楚miR-135b-5p与CRC中西妥昔单抗(CTx)耐药是否存在相关性.使用GEO数据库测量miR-135b-5p在CRC中的表达。此外,应用RT-qPCR来确定miR-135b-5p在三种人CRC细胞和NCM460细胞中的产生水平。利用伤口愈合和transwell测定法检查细胞迁移和侵入的能力,虽然CCK-8测定用于评估细胞活力,以及增殖的集落形成测定。已经使用蛋白质印迹研究了CRC细胞西妥昔单抗抗性中miR-135b-5p的预期靶蛋白。抑制miR-135b-5p可提高CTx耐药CRC细胞的CTx敏感性,如增殖减弱所示,迁移,和入侵能力。机制研究显示miR-135b-5p通过下调FOXN3调节上皮-间质转化(EMT)过程和Wnt/β-catenin信号通路。总之,敲除miR-135b-5p可以增加CRC细胞中FOXN3的表达,推进EMT流程,同时激活Wnt/β-catenin信号通路提高CRC细胞的CTx抗性。
    Despite advances in targeted therapies, primary and acquired resistance make the treatment of colorectal cancer (CRC) a pressing issue to be resolved. According to reports, the development of CRC is linked to miRNA dysregulation. Multiple studies have demonstrated that miR-135b-5p has an aberrant expression level between CRC tissues and adjacent tissues. However, it is unclear whether there is a correlation between miR-135b-5p and cetuximab (CTx) resistance in CRC. Use the GEO database to measure miR-135b-5p expression in CRC. Additionally, RT-qPCR was applied to ascertain the production level of miR-135b-5p in three human CRC cells and NCM460 cells. The capacity of cells to migrate and invade was examined utilizing the wound-healing and transwell assays, while the CCK-8 assay served for evaluating cell viability, as well as colony formation assays for proliferation. The expected target protein of miR-135b-5p in CRC cell cetuximab resistance has been investigated using western blot. Suppression of miR-135b-5p could increase the CTx sensitivity of CTx-resistant CRC cells, as manifested by the attenuation of proliferation, migration, and invasion ability. Mechanistic studies revealed miR-135b-5p regulates the epithelial-to-mesenchymal transition (EMT) process and Wnt/β-catenin signaling pathway through downgulating FOXN3. In short, knockdowning miR-135b-5p could increase FOXN3 expression in CRC cells, promote the EMT process, and simultaneously activate the Wnt/β-catenin signaling pathway to elevate CTx resistance in CRC cells.
    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
    氨基酸是蛋白质的重要组成部分,细胞生存的关键能源,和支持肿瘤细胞抗性生长的关键信号分子。在肿瘤细胞中,氨基酸代谢重编程的特点是氨基酸的摄取增强以及它们的异常合成,击穿,和运输,导致免疫逃避和肿瘤细胞的恶性进展。本文综述了肿瘤细胞中氨基酸代谢的改变及其对肿瘤微环境的影响。并概述了目前氨基酸代谢的临床应用。针对氨基酸代谢的创新药物在精准和个性化癌症治疗方面具有广阔的前景。
    Amino acids are essential building blocks for proteins, crucial energy sources for cell survival, and key signaling molecules supporting the resistant growth of tumor cells. In tumor cells, amino acid metabolic reprogramming is characterized by the enhanced uptake of amino acids as well as their aberrant synthesis, breakdown, and transport, leading to immune evasion and malignant progression of tumor cells. This article reviews the altered amino acid metabolism in tumor cells and its impact on tumor microenvironment, and also provides an overview of the current clinical applications of amino acid metabolism. Innovative drugs targeting amino acid metabolism hold great promise for precision and personalized cancer 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
    治疗肿瘤患者的一个相关挑战是对化疗的耐药性的发展。免疫-,和放射治疗。尽管人们对治疗抵抗的原因知之甚少,有证据表明,它依赖于代偿机制,即细胞发育以取代特定的细胞内信号,这些信号在药物抑制后应该是无活性的。其中一种机制涉及整合素,连接细胞与细胞外基质并在细胞迁移中起关键作用的膜受体。一种特定类型的整合素的阻断通常由另一种整合素二聚体的过表达来补偿。通常支持细胞粘附和迁移。特别是,整合素αvβ3是参与肿瘤对酪氨酸激酶抑制剂治疗耐药的关键受体,免疫检查点抑制剂,和放射治疗;然而,αvβ3整联蛋白的特异性抑制不足以避免肿瘤复发。这里,我们回顾了整合素αvβ3在肿瘤对治疗的抵抗中的作用以及迄今为止已经提出的机制。尽管我们关注的是αvβ3整合素,值得注意的是,其他整合素也与耐药性有关,这些受体之间的协同作用不可忽视。
    A relevant challenge for the treatment of patients with neoplasia is the development of resistance to chemo-, immune-, and radiotherapies. Although the causes of therapy resistance are poorly understood, evidence suggests it relies on compensatory mechanisms that cells develop to replace specific intracellular signaling that should be inactive after pharmacological inhibition. One such mechanism involves integrins, membrane receptors that connect cells to the extracellular matrix and have a crucial role in cell migration. The blockage of one specific type of integrin is frequently compensated by the overexpression of another integrin dimer, generally supporting cell adhesion and migration. In particular, integrin αvβ3 is a key receptor involved in tumor resistance to treatments with tyrosine kinase inhibitors, immune checkpoint inhibitors, and radiotherapy; however, the specific inhibition of the αvβ3 integrin is not enough to avoid tumor relapse. Here, we review the role of integrin αvβ3 in tumor resistance to therapy and the mechanisms that have been proposed thus far. Despite our focus on the αvβ3 integrin, it is important to note that other integrins have also been implicated in drug resistance and that the collaborative action between these receptors should not be neglected.
    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
    胶质母细胞瘤(GBM)是一种极具侵袭性的脑肿瘤,生存率低。目前的治疗方法,如化疗,辐射,由于肿瘤生长,手术是有问题的,入侵,和肿瘤微环境。GBM细胞对这些标准治疗有抵抗力,而肿瘤的异质性使得很难找到一种通用的办法。GBM的进展和对治疗的抗性的获得是由于肿瘤细胞和TME之间复杂的相互作用。TME的很大一部分由炎症浸润组成,小胶质细胞和巨噬细胞是主要细胞。
    对5年的文献数据的分析表明,肿瘤相关巨噬细胞(TAMs)能够释放促进肿瘤增殖的细胞因子和生长因子,生存,和转移,同时抑制免疫细胞功能。
    因此,免疫抑制状态,提供这种深入研究的TME细胞,应该通过TAMs调节肿瘤治疗抗性和侵袭性来促进GBM的发展。因此,TAM是治疗胶质母细胞瘤的一个有吸引力的治疗靶点。
    这篇综述全面概述了有关TAM性质的最新研究以及针对TAM的治疗策略的发展,专注于巨噬细胞特性的多样性,被调制,以及分子靶标。
    UNASSIGNED: Glioblastoma (GBM) is an extremely aggressive form of brain tumor with low survival rates. Current treatments such as chemotherapy, radiation, and surgery are problematic due to tumor growth, invasion, and tumor microenvironment. GBM cells are resistant to these standard treatments, and the heterogeneity of the tumor makes it difficult to find a universal approach. Progression of GBM and acquisition of resistance to therapy are due to the complex interplay between tumor cells and the TME. A significant portion of the TME consists of an inflammatory infiltrate, with microglia and macrophages being the predominant cells.
    UNASSIGNED: Analysis of the literature data over a course of 5 years suggest that the tumor-associated macrophages (TAMs) are capable of releasing cytokines and growth factors that promote tumor proliferation, survival, and metastasis while inhibiting immune cell function at the same time.
    UNASSIGNED: Thus, immunosuppressive state, provided with this intensively studied kind of TME cells, is supposed to promote GBM development through TAMs modulation of tumor treatment-resistance and aggressiveness. Therefore, TAMs are an attractive therapeutic target in the treatment of glioblastoma.
    UNASSIGNED: This review provides a comprehensive overview of the latest research on the nature of TAMs and the development of therapeutic strategies targeting TAMs, focusing on the variety of macrophage properties, being modulated, as well as molecular targets.
    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
    第二代雄激素受体(AR)信号抑制剂(ARSI),如阿比特龙和恩扎鲁他胺,延长去势抵抗性前列腺癌(CRPC)患者的寿命。然而,接受ARSI的患者最终会通过各种复杂的机制产生耐药性,包括AR突变,组成型活性AR-剪接变体(AR-Vs),AR过度表达。这里,我们表征了一种新型的AR纯拮抗剂,TAS3681抑制AR转录活性并下调AR全长(AR-FL)和AR-Vs。TAS3681降低了恩杂鲁胺抗性细胞中AR-FL和AR-Vs(SASMDVNo.3-14),在体外和体内,在AR-V7阳性异种移植模型中显示强的抗肿瘤功效。在过表达AR的VCaP(前列腺癌)细胞中,相反,恩扎鲁他胺,TAS3681有效抑制细胞增殖并下调AR表达。重要的是,TAS3681阻断了各种突变ARs的转录活性,包括突变F877L/T878A和H875Y/T878A,赋予恩扎鲁他胺抗性,和V716M和H875Y突变,赋予达洛鲁胺抗性。我们的结果表明,TAS3681抑制AR信号的再激活,导致对ARSI的抵抗,通过新确定的作用机制。因此,TAS3681可能是CRPC治疗的一种新的治疗选择。
    Second-generation androgen receptor (AR) signaling inhibitors (ARSIs), such as abiraterone and enzalutamide, prolong the life of patients with castration-resistant prostate cancer (CRPC). However, patients receiving ARSIs ultimately develop resistance through various complex mechanisms, including AR mutations, constitutively active AR-splice variants (AR-Vs), and AR overexpression. Here, we characterized a novel AR pure antagonist, TAS3681, which inhibits AR transcriptional activity and downregulates AR-full length (AR-FL) and AR-Vs. TAS3681 reduced the protein levels of AR-FL and AR-Vs including AR-V7 in enzalutamide-resistant cells (SAS MDV No. 3-14), in vitro and in vivo, showing strong antitumor efficacy in an AR-V7-positive xenograft model. In AR-overexpressing VCaP (prostate cancer) cells, conversely to enzalutamide, TAS3681 effectively suppressed cell proliferation and downregulated AR expression. Importantly, TAS3681 blocked the transcriptional activity of various mutant ARs, including mutations F877L/T878A and H875Y/T878A, which confer resistance to enzalutamide, and V716M and H875Y mutations, which confer resistance to darolutamide. Our results demonstrate that TAS3681 suppresses the reactivation of AR signaling, which causes resistance to ARSIs, via a newly identified mechanism of action. Therefore, TAS3681 could be a new therapeutic option for CRPC 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: 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
    鼻咽癌(NPC),作为头颈部最常见的恶性肿瘤之一,对其发病机制仍缺乏完整的了解。目前,放射治疗,同步放化疗,靶向治疗是治疗NPC的主要方法。随着医学的进步,鼻咽癌的治愈率一直在稳步上升。然而,复发和转移是治疗失败的主要原因。因此,对鼻咽癌发生和发展的分子机制进行了深刻的探索,随着相应治疗方法的探索,在寻求与这种疾病作斗争的全面解决方案时变得尤为必要。高迁移率组AT-hook2(HMGA2)是一种能够改变染色质结构的关键蛋白,调节基因表达,并影响转录活性。在癌症研究领域,HMGA2表现出广泛的失调,在几乎所有的恶性肿瘤中起着至关重要的作用。它与各种致瘤过程有关,包括细胞周期调节,细胞增殖,上皮-间质转化,血管生成,肿瘤侵袭,转移,和抗药性。此外,HMGA2在某些恶性肿瘤中作为分子标志物和独立的预后因素。最近的研究越来越揭示了HMGA2在鼻咽癌(NPC)中的关键作用,特别是在促进恶性进展方面,与肿瘤耐药性相关,并作为独立的不良预后因素。本文就HMGA2在鼻咽癌中的致癌作用作一综述。提示其与鼻咽癌化疗耐药的潜在关联,并提出其候选资格是鼻咽癌预后评估的独立因素。
    Nasopharyngeal carcinoma (NPC), as one of the most prevalent malignancies in the head and neck region, still lacks a complete understanding of its pathogenesis. Presently, radiotherapy, concurrent chemoradiotherapy, and targeted therapy stand as the primary modalities for treating NPC. With advancements in medicine, the cure rates for nasopharyngeal carcinoma have been steadily increasing. Nevertheless, recurrence and metastasis persist as the primary reasons for treatment failure. Consequently, a profound exploration of the molecular mechanisms underlying the occurrence and progression of nasopharyngeal carcinoma, along with the exploration of corresponding therapeutic approaches, becomes particularly imperative in the quest for comprehensive solutions to combat this disease. High mobility group AT-hook 2 (HMGA2) is a pivotal protein capable of altering chromatin structure, regulating gene expression, and influencing transcriptional activity. In the realm of cancer research, HMGA2 exhibits widespread dysregulation, playing a crucial role in nearly all malignant tumors. It is implicated in various tumorigenic processes, including cell cycle regulation, cell proliferation, epithelial-mesenchymal transition, angiogenesis, tumor invasion, metastasis, and drug resistance. Additionally, HMGA2 serves as a molecular marker and an independent prognostic factor in certain malignancies. Recent studies have increasingly unveiled the critical role of HMGA2 in nasopharyngeal carcinoma (NPC), particularly in promoting malignant progression, correlating with tumor resistance, and serving as an independent adverse prognostic factor. This review focuses on elucidating the oncogenic role of HMGA2 in NPC, suggesting its potential association with chemotherapy resistance in NPC, and proposing its candidacy as an independent factor in nasopharyngeal carcinoma prognosis assessment.
    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
    全世界的癌症发病率和死亡人数一直在增加。然而,常规疗法有一些明显的局限性,例如非特异性靶向,全身性毒性作用,尤其是肿瘤的多药耐药性(MDR),其中,自噬起着至关重要的作用。因此,迫切需要新的治疗方法来减少不良反应,提高治疗效果,更有效、准确地扩大其治疗适应证。基于自噬调节因子的联合治疗是克服肿瘤耐药和提高抗肿瘤药物敏感性的一种非常可行和重要的方法。然而,功效改善越少,更多的全身毒性等问题限制了其临床应用。纳米技术提供了克服这一限制的好方法。自噬调节因子与抗肿瘤药物通过纳米平台共同递送,为肿瘤的治疗提供了良好的治疗策略,尤其是耐药肿瘤。值得注意的是,具有自噬调控特性的纳米材料作为载体平台具有广阔的治疗前景,尤其是辅助治疗。然而,仍然需要进一步的研究来克服诸如安全,生物相容性,和纳米医学的副作用。此外,临床研究对于证实其在肿瘤治疗中的应用也是必不可少的。
    The overall cancer incidence and death toll have been increasing worldwide. However, the conventional therapies have some obvious limitations, such as non-specific targeting, systemic toxic effects, especially the multidrug resistance (MDR) of tumors, in which, autophagy plays a vital role. Therefore, there is an urgent need for new treatments to reduce adverse reactions, improve the treatment efficacy and expand their therapeutic indications more effectively and accurately. Combination therapy based on autophagy regulators is a very feasible and important method to overcome tumor resistance and sensitize anti-tumor drugs. However, the less improved efficacy, more systemic toxicity and other problems limit its clinical application. Nanotechnology provides a good way to overcome this limitation. Co-delivery of autophagy regulators combined with anti-tumor drugs through nanoplatforms provides a good therapeutic strategy for the treatment of tumors, especially drug-resistant tumors. Notably, the nanomaterials with autophagy regulatory properties have broad therapeutic prospects as carrier platforms, especially in adjuvant therapy. However, further research is still necessary to overcome the difficulties such as the safety, biocompatibility, and side effects of nanomedicine. In addition, clinical research is also indispensable to confirm its application in tumor treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号