tumor resistance

肿瘤耐药
  • 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: 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)

  • 文章类型: Journal Article
    具核梭杆菌,一种主要存在于口腔中的厌氧革兰氏阴性细菌,因其在癌症进展和预后中的新兴作用而受到关注。虽然广泛的研究揭示了核梭杆菌和结直肠癌之间的机械联系,关于其在结直肠癌以外的癌症中的存在和转移影响的全面审查显然缺乏。本文将我们的观点从结直肠癌扩展到与核梭杆菌相关的各种恶性肿瘤,包括口服,胰腺,食道,乳房,和胃癌。我们的中心重点是解开管核梭杆菌定植的机制,initiation,以及促进不同癌症类型的转移。此外,我们探讨了核梭杆菌对癌症治疗的不利影响,特别是在免疫治疗和化疗领域。此外,本文强调核梭杆菌作为一种潜在的肿瘤生物标志物和治疗靶点的临床研究意义,对其在癌症检测和预后评估中的适用性提出了新的展望。
    Fusobacterium nucleatum, an anaerobic Gram-negative bacterium primarily residing in the oral cavity, has garnered significant attention for its emerging role in cancer progression and prognosis. While extensive research has revealed mechanistic links between Fusobacterium nucleatum and colorectal cancer, a comprehensive review spanning its presence and metastatic implications in cancers beyond colorectal origin is conspicuously absent. This paper broadens our perspective from colorectal cancer to various malignancies associated with Fusobacterium nucleatum, including oral, pancreatic, esophageal, breast, and gastric cancers. Our central focus is to unravel the mechanisms governing Fusobacterium nucleatum colonization, initiation, and promotion of metastasis across diverse cancer types. Additionally, we explore Fusobacterium nucleatum\'s adverse impacts on cancer therapies, particularly within the domains of immunotherapy and chemotherapy. Furthermore, this paper underscores the clinical research significance of Fusobacterium nucleatum as a potential tumor biomarker and therapeutic target, offering a novel outlook on its applicability in cancer detection and prognostic 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
    肿瘤耐药机制复杂,可能涉及干细胞维持,上皮-间质转化,存活信号通路的激活,转运蛋白表达,和肿瘤微环境重塑,所有这些都与γ-分泌酶/Notch信号连接。越来越多的证据表明,γ-分泌酶/Notch途径的激活是癌症进展和耐药性发展的关键驱动因素,γ-分泌酶抑制剂(GSI)可能是通过靶向逆转肿瘤化疗耐药性的最有希望的药物。γ-分泌酶/Notch途径。这里,我们系统地总结了支持γ-分泌酶/Notch激活相关的癌细胞转化为癌症干细胞的作用,EMT流程的推广,PI3K/Akt,MEK/ERK和NF-κB激活,ABC转运蛋白表达的增强,和TME改变介导肿瘤耐药。随后,我们分析了GSIs靶向γ-分泌酶/Notch通路逆转肿瘤耐药的机制,并提出了GSIs在治疗乳腺癌耐药方面优于其他肿瘤的突出优势。最后,我们强调,用于逆转肿瘤耐药性的GSI的开发是有希望的。
    The mechanism of tumor drug resistance is complex and may involve stem cell maintenance, epithelial-mesenchymal transition, the activation of survival signaling pathways, transporter protein expression, and tumor microenvironment remodeling, all of which are linked to γ-secretase/Notch signaling. Increasing evidence has shown that the activation of the γ-secretase/Notch pathway is a key driver of cancer progression and drug resistance development and that γ-secretase inhibitors (GSIs) may be the most promising agents for reversing chemotherapy resistance of tumors by targeting the γ-secretase/Notch pathway. Here, we systematically summarize the roles in supporting γ-secretase/Notch activation-associated transformation of cancer cells into cancer stem cells, promotion of the EMT process, PI3K/Akt, MEK/ERK and NF-κB activation, enhancement of ABC transporter protein expression, and TME alteration in mediating tumor drug resistance. Subsequently, we analyze the mechanism of GSIs targeting the γ-secretase/Notch pathway to reverse tumor drug resistance and propose the outstanding advantages of GSIs in treating breast cancer drug resistance over other tumors. Finally, we emphasize that the development of GSIs for reversing tumor drug resistance is promising.
    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: Case Reports
    人表皮生长因子受体2(HER2)阳性乳腺癌由于HER2基因过表达/扩增而与较高的转移风险和较差的总生存期(OS)相关。尽管抗HER2靶向治疗在HER2阳性晚期乳腺癌(ABC)患者中显示出生存益处,长期治疗往往会导致耐药性,使进一步的治疗选择复杂化。RC48,一种抗体-药物偶联物(ADC),结合了抗体靶向的好处和小分子药物的细胞毒性作用。
    我们介绍了一例HER2阳性ABC的女性患者,该患者在多线抗HER2靶向治疗后出现耐药性和疾病进展。在这种情况下,RC48在对HER2靶向治疗有抗性的ABC患者中表现出抗肿瘤活性。在使用120mgRC48的八个治疗周期后,肿瘤大小减小并稳定。
    本病例报告强调了RC48作为HER2靶向治疗耐药患者的潜在临床价值。
    UNASSIGNED: Human epidermal growth factor receptor 2 (HER2)-positive breast cancer is associated with a higher risk of metastasis and poorer overall survival (OS) due to HER2 gene overexpression/amplification. Although anti-HER2 targeted therapy has shown survival benefits in HER2-positive advanced breast cancer (ABC) patients, long-term treatment often leads to drug resistance, complicating further treatment options. RC48, an antibody-drug conjugate (ADC), combines the benefits of antibody targeting with the cytotoxic effects of a small molecule drug.
    UNASSIGNED: We present a case involving a female patient with HER2-positive ABC who developed drug resistance and disease progression following multi-line anti-HER2 targeted therapy. In this instance, RC48 exhibited anti-tumor activity in an ABC patient resistant to HER2-targeted therapy. After eight treatment cycles with 120 mg of RC48, the tumor size decreased and stabilized.
    UNASSIGNED: This case report underscores the potential clinical value of RC48 as a promising treatment alternative for patients resistant to HER2 targeted 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: 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)

公众号