Gastrointestinal cancers

胃肠道肿瘤
  • 文章类型: Journal Article
    目的:本研究系统评价三级淋巴结构(TLS)与胃肠道肿瘤临床病理特征及免疫浸润细胞的关系。
    方法:我们搜索了Webofscience,Pubmed,Embase,和Cochrane图书馆进行截至2023年7月1日满足要求的研究,以及赔率比,相应的95%置信区间或均值和标准差,包括在分析中。
    结果:我们最终纳入了20项研究,共涉及4856名患者。发现TLS与T期显著相关,N级,TNM阶段,和肿瘤大小。此外,TLS阳性的患者显示T细胞相关标志物的表达显着升高,包括CD3,CD4,CD8,CD45RO;B细胞相关标志物,例如CD11c和CD20;以及树突状细胞相关标志物CD103。另一方面,TLS阳性与FOXP3和CD68的低表达显著相关。此外,TLS与肿瘤浸润淋巴细胞的整体浸润之间存在显著正相关.
    结论:在胃肠道肿瘤中,TLS的存在与各种免疫细胞的浸润显著相关。为了确定成熟TLS的存在和适当的免疫细胞浸润之间的理想平衡,需要进一步开展高质量和多中心的临床研究.
    OBJECTIVE: This study systematically evaluated the relationship between tertiary lymphoid structures (TLS) and clinical pathological features as well as immune infiltrating cells in gastrointestinal cancers.
    METHODS: We searched Web of science, Pubmed, Embase, and Cochrane Library for studies that met the requirements as of July 1, 2023, and the odds ratio, the corresponding 95% confidence interval or mean and standard deviation, were included in the analysis.
    RESULTS: We eventually included 20 studies, involving a total of 4856 patients. TLS were found to be significantly associated with T stage, N stage, TNM stage, and tumor size. Moreover, patients with positive TLS showed significantly elevated expression of T-cell related markers, including CD3, CD4, CD8, CD45RO; B-cell related markers, such as CD11c and CD20; and dendritic cell-related marker CD103. On the other hand, positive TLS correlated significantly with low expression of FOXP3 and CD68. Additionally, there was a significant positive association between TLS and overall infiltration of tumor-infiltrating lymphocytes.
    CONCLUSIONS: The presence of TLS is significantly correlated with the infiltration of various immune cells in gastrointestinal cancers. To determine the ideal balance between the presence of mature TLS and appropriate immune cell infiltration, further high-quality and multicenter clinical studies need to be conducted.
    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
    胃肠道(GI)癌症,包括消化道内的一系列恶性肿瘤,在诊断和治疗方面都存在重大挑战,反映了对创新治疗策略的迫切需要。本文探讨了非组蛋白甲基化对胃肠道(GI)癌症的发病机制和进化的深远影响。非组蛋白,通过蛋白质精氨酸甲基转移酶(PRMTs)和赖氨酸甲基转移酶(KMTs)等酶进行甲基化,在细胞信号传导中起关键作用,新陈代谢,染色质重塑,和其他对癌症发展至关重要的过程。这篇综述阐明了非组蛋白甲基化影响肿瘤生物学关键方面的复杂机制。包括肿瘤发生,增长,扩散,入侵,迁移,代谢重编程,和胃肠道恶性肿瘤的免疫逃逸。突出了最近的发现,这项工作强调了非组蛋白甲基化在癌症生物学中的重要性,以及其作为旨在改善胃肠道癌症患者结局的创新治疗策略靶标的潜力.
    Gastrointestinal (GI) cancers, encompassing a range of malignancies within the digestive tract, present significant challenges in both diagnosis and treatment, reflecting a dire need for innovative therapeutic strategies. This article delves into the profound influence of non-histone methylation on the pathogenesis and evolution of gastrointestinal (GI) cancers. Non-histone proteins, undergoing methylation by enzymes such as Protein Arginine Methyltransferases (PRMTs) and Lysine Methyltransferases (KMTs), play pivotal roles in cellular signaling, metabolism, chromatin remodeling, and other processes crucial for cancer development. This review illuminates the complex mechanisms by which non-histone methylation affects key aspects of tumor biology, including oncogenesis, growth, proliferation, invasion, migration, metabolic reprogramming, and immune escape in GI malignancies. Highlighting recent discoveries, this work underscores the importance of non-histone methylation in cancer biology and its potential as a target for innovative therapeutic strategies aimed at improving outcomes for patients with GI cancers.
    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
    胃肠道是大多数肠道微生物群沉降的地方;因此,肠道微生物群的组成和代谢物的变化,以及它们对免疫系统的调节作用,对胃肠道疾病的发展有非常重要的影响。本文旨在综述肠道菌群在宿主环境和免疫代谢系统中的作用,并总结植物活性成分对胃肠道肿瘤的有益作用。从而为胃肠道疾病的预防和治疗提供前瞻性见解。在PubMed数据库上进行了文献检索,关键字为“胃肠道癌”,“肠道微生物群”,“免疫代谢”,\"SCFA\",“胆汁酸”,\"多胺\",\"色氨酸\",“细菌素”,“免疫细胞”,“能量代谢”,\"多酚\",“多糖”,“生物碱”,和“三萜”。肠道菌群组成的变化影响了胃肠道疾病,而它们的代谢物,例如SCFA,细菌素,和植物代谢产物,可以阻止胃肠道癌症和多胺-,色氨酸-,和胆汁酸诱导的致癌机制。GPRC,HDAC,FXRs,和AHR是影响胃肠道肿瘤发生发展的肠道微生物代谢产物的重要受体信号。植物活性成分通过影响肠道微生物的组成和调节免疫代谢对胃肠道肿瘤产生积极影响。胃肠道肿瘤可以通过改变肠道微生物环境来改善,施用植物活性成分进行治疗,刺激或阻断免疫代谢信号分子。尽管对微生物群的研究越来越广泛,与预防胃肠道疾病的自主致病因素相比,它似乎更多地代表了与充足纤维摄入相关的肠道健康状况指标。本研究详细介绍了胃肠道肿瘤的发病机制和用于治疗的植物活性成分,希望为更简单的研究提供灵感,更安全,以及本领域更有效的治疗途径或治疗剂。
    The gastrointestinal tract is where the majority of gut microbiota settles; therefore, the composition of the gut microbiota and the changes in metabolites, as well as their modulatory effects on the immune system, have a very important impact on the development of gastrointestinal diseases. The purpose of this article was to review the role of the gut microbiota in the host environment and immunometabolic system and to summarize the beneficial effects of botanical active ingredients on gastrointestinal cancer, so as to provide prospective insights for the prevention and treatment of gastrointestinal diseases. A literature search was performed on the PubMed database with the keywords \"gastrointestinal cancer\", \"gut microbiota\", \"immunometabolism\", \"SCFAs\", \"bile acids\", \"polyamines\", \"tryptophan\", \"bacteriocins\", \"immune cells\", \"energy metabolism\", \"polyphenols\", \"polysaccharides\", \"alkaloids\", and \"triterpenes\". The changes in the composition of the gut microbiota influenced gastrointestinal disorders, whereas their metabolites, such as SCFAs, bacteriocins, and botanical metabolites, could impede gastrointestinal cancers and polyamine-, tryptophan-, and bile acid-induced carcinogenic mechanisms. GPRCs, HDACs, FXRs, and AHRs were important receptor signals for the gut microbial metabolites in influencing the development of gastrointestinal cancer. Botanical active ingredients exerted positive effects on gastrointestinal cancer by influencing the composition of gut microbes and modulating immune metabolism. Gastrointestinal cancer could be ameliorated by altering the gut microbial environment, administering botanical active ingredients for treatment, and stimulating or blocking the immune metabolism signaling molecules. Despite extensive and growing research on the microbiota, it appeared to represent more of an indicator of the gut health status associated with adequate fiber intake than an autonomous causative factor in the prevention of gastrointestinal diseases. This study detailed the pathogenesis of gastrointestinal cancers and the botanical active ingredients used for their treatment in the hope of providing inspiration for research into simpler, safer, and more effective treatment pathways or therapeutic agents in the field.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    针对claudin-18亚型2(CLDN18.2)的多种疗法正在开发中,用于治疗晚期胃食管腺癌和其他实体瘤。在2024年美国临床肿瘤学会(ASCO)年会上,介绍了SPOTLIGHT3期试验的最终结果,证明在CLDN18.2表达≥75%的晚期胃食管腺癌的一线治疗中,在化疗中加入CLDN18.2特异性抗体唑贝妥昔单抗可显著获益生存.ASCO2024发表的早期试验结果显示,去岩藻糖基化CLDN18.2特异性抗体FG-M108与化疗联合治疗CLDN18.2阳性晚期胃食管癌和胰腺癌的一线治疗具有良好的疗效和安全性。此外,ASCO2024上发表的几项早期试验研究了CLDN18.2阳性难治性晚期实体瘤的其他CLDN18.2靶向方法,包括CLDN18.2靶向抗体-药物缀合物LM-302和IBI343,双特异性抗CLDN18.2/CD3抗体IBI38和嵌合抗原受体T细胞疗法。这些新方法可能会将CLDN18.2靶向疗法的益处扩展到更广泛的肿瘤类型和表达较低水平CLDN18.2的肿瘤。
    Multiple classes of therapies targeting claudin-18 isoform 2 (CLDN18.2) are under development for the treatment of advanced gastroesophageal adenocarcinoma and other solid tumors. At the 2024 American Society of Clinical Oncology (ASCO) Annual Meeting, the final results of the phase 3 SPOTLIGHT trial were presented, demonstrating a significant survival benefit from the addition of the CLDN18.2-specific antibody zolbetuximab to chemotherapy in the first-line treatment of advanced gastroesophageal adenocarcinomas with ≥ 75% CLDN18.2 expression. Early-phase trial results presented at ASCO 2024 showed promising efficacy and safety of the afucosylated CLDN18.2-specific antibody FG-M108 in combination with chemotherapy in the first-line treatment of CLDN18.2-positive advanced gastroesophageal and pancreatic cancers. In addition, several early-phase trials presented at ASCO 2024 investigate other CLDN18.2-targeting approaches in CLDN18.2-positive refractory advanced solid tumors, including the CLDN18.2-targeting antibody-drug conjugates LM-302 and IBI343, the bispecific anti-CLDN18.2/CD3 antibody IBI38, and the chimeric antigen receptor T cell therapy satricabtagene autoleucel. These novel approaches could potentially expand the benefit of CLDN18.2-targeting therapies to a broader range of tumor types and to tumors expressing lower levels of CLDN18.2.
    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
    代谢功能障碍相关的脂肪变性肝病(MASLD)在全球范围内造成了显着且不断增加的健康和经济负担。大量流行病学证据表明,MASLD是一种多系统疾病,不仅与肝脏相关的并发症有关,而且与发生心脏代谢合并症和肝外癌症(主要是胃肠道[GI]癌症)的风险增加有关。胃肠道癌症占全球癌症发病率的四分之一,占癌症相关死亡人数的三分之一。在这篇叙述性评论中,我们提供了有关(a)MASLD中非肝胃肠道癌风险的流行病学数据的文献综述,(b)MASLD(以及与MASLD相关的因素)可能增加这种风险的假定机制,和(c)可能的药物疗法有益地影响MASLD和肝外GI癌风险。有多种潜在的病理生理机制,MASLD可能增加肝外胃肠道癌的风险。虽然还需要进一步的研究,目前的证据支持MASLD可能的肝外致癌作用,无论肥胖和糖尿病的状态,从而突出了为MASLD患者量身定制癌症筛查的潜在作用。尽管文献中有相互矛盾的数据,阿司匹林,他汀类药物和二甲双胍似乎对胃肠道癌具有一定的化学预防作用。
    Metabolic dysfunction-associated steatotic liver disease (MASLD) poses a significant and ever-increasing health and economic burden worldwide. Substantial epidemiological evidence shows that MASLD is a multisystem disease that is associated not only with liver-related complications but is also associated with an increased risk of developing cardiometabolic comorbidities and extrahepatic cancers (principally gastrointestinal [GI] cancers). GI cancers account for a quarter of the global cancer incidence and a third of cancer-related deaths. In this narrative review, we provide an overview of the literature on (a) the epidemiological data on the risk of non-liver GI cancers in MASLD, (b) the putative mechanisms by which MASLD (and factors linked with MASLD) may increase this risk, and (c) the possible pharmacotherapies beneficially affecting both MASLD and extrahepatic GI cancer risk. There are multiple potential pathophysiological mechanisms by which MASLD may increase extrahepatic GI cancer risk. Although further studies are needed, the current evidence supports a possible extrahepatic carcinogenic role for MASLD, regardless of obesity and diabetes status, thus highlighting the potential role of tailoring cancer screening for individuals with MASLD. Although there are conflicting data in the literature, aspirin, statins and metformin appear to exert some chemo-preventive effects against GI cancer.
    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
    p53是一种关键的肿瘤抑制因子,其正常功能的破坏通常是肿瘤发展或进展的先决条件。我们以前的工作表明,Krüppel相关盒(KRAB)域锌指蛋白(KZFPs)家族的多个成员通过与p53相互作用来调节p53的转录活性。但是这些成员的肿瘤生物学功能尚未完全阐明。这里,与胃肠道癌症(GIC)相关的泛癌症分析显示,p53相互作用蛋白ZNF8,是恶性肿瘤患者的不良预后因素。ZNF8与p53相互作用并进一步抑制其在结肠癌细胞中的转录活性。ZNF8的敲低或ZNF8的过表达抑制或促进体外集落形成,迁移,入侵,和p53+/+结肠癌HCT116细胞的血管生成,HepG2细胞和EC109细胞而不是p53-/-结肠癌HCT116细胞和p53敲除的HepG2细胞,分别。在体内进行的异种移植实验还显示,p53+/+而p53-/-HCT116细胞中ZNF8的敲低抑制了肿瘤的生长和向肺的转移,延长荷瘤小鼠的寿命。临床上,结肠癌和食管癌的两个独立的免疫组织化学队列还表明,ZNF8在癌组织中的表达高于癌旁组织,这与无p53突变患者的总体生存结局较差相关.一起,我们的结果为ZNF8的p53特异性肿瘤致癌功能提供了见解。ZNF8可能被证明是GIC治疗的潜在靶标。
    p53 is a critical tumor suppressor, and the disruption of its normal function is often a prerequisite for the development or progression of tumors. Our previous works revealed that multiple members of Krüppel-associated box (KRAB) domain zinc-finger proteins (KZFPs) family regulate p53 transcriptional activity by interacting with it. But the tumor biology functions of these members have not been fully elucidated. Here, the pan-cancer analysis related to gastrointestinal cancers (GICs) revealed that ZNF8, a p53-interacting protein, is an unfavorable prognostic factor for patients with malignancies. ZNF8 interacts with p53 and further depresses its transcriptional activity in colon cancer cells. The knockdown of ZNF8 or the overexpression of ZNF8 inhibits or facilitates the in vitro colony formation, migration, invasion, and angiogenesis of p53+/+ colon cancer HCT116 cells, HepG2 cells and EC109 cells rather than p53-/- colon cancer HCT116 cells and p53-knockout HepG2 cells, respectively. Xenograft experiments conducted in vivo also showed that the knockdown of ZNF8 in p53+/+ but not in p53-/- HCT116 cells curbs the tumor growth and metastasis to lung, leading to an extended life span for tumor-bearing mice. Clinically, two independent immunohistochemistry cohorts of colon cancer and esophageal cancer also indicated that ZNF8 is higher expression in carcinoma tissues than adjacent tissues and this is associated with worse overall survival outcomes in patients without harboring p53 mutation. Together, our results provide insight into the p53-specific tumor oncogenic function of ZNF8. ZNF8 may prove to be a potential target for GICs treatment.
    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
    胃肠道(GI)癌症代表了重大的全球健康挑战,推动不懈的努力,以确定创新的诊断和治疗方法。微生物组研究的最新进展揭示了以前被低估的癌症进展维度,该维度围绕胃肠道癌症和宿主肠道微生物群之间复杂的代谢相互作用。这篇综述旨在全面概述这些新兴的代谢相互作用及其在胃肠道癌症的精确诊断和治疗突破中催化范式转变的潜力。本文通过深入研究宿主代谢与肠道微生物群之间的共生关系,强调了微生物组研究对肿瘤学的开创性影响。它提供了为个体患者量身定制治疗策略的宝贵见解,从而超越了传统的一刀切的方法。这篇综述还揭示了新的诊断方法,可以改变胃肠道癌症的早期检测。可能导致更有利的患者结果。总之,探索宿主肠道微生物群和胃肠道癌症之间的代谢相互作用,展示了在对抗这些强大疾病的持续斗争中一个有希望的前沿.通过理解和利用微生物组的影响,对胃肠道癌症的精准诊断和治疗创新的未来似乎更加乐观,为量身定制的治疗方法和提高诊断精度打开门。
    Gastrointestinal (GI) cancers represent a significant global health challenge, driving relentless efforts to identify innovative diagnostic and therapeutic approaches. Recent strides in microbiome research have unveiled a previously underestimated dimension of cancer progression that revolves around the intricate metabolic interplay between GI cancers and the host\'s gut microbiota. This review aims to provide a comprehensive overview of these emerging metabolic interactions and their potential to catalyze a paradigm shift in precision diagnosis and therapeutic breakthroughs in GI cancers. The article underscores the groundbreaking impact of microbiome research on oncology by delving into the symbiotic connection between host metabolism and the gut microbiota. It offers valuable insights into tailoring treatment strategies to individual patients, thus moving beyond the traditional one-size-fits-all approach. This review also sheds light on novel diagnostic methodologies that could transform the early detection of GI cancers, potentially leading to more favorable patient outcomes. In conclusion, exploring the metabolic interactions between host gut microbiota and GI cancers showcases a promising frontier in the ongoing battle against these formidable diseases. By comprehending and harnessing the microbiome\'s influence, the future of precision diagnosis and therapeutic innovation for GI cancers appears more optimistic, opening doors to tailored treatments and enhanced diagnostic precision.
    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
    胃肠道癌症(GIC)是威胁全球人类健康的高度流行的癌症。据报道,Wnt/β-catenin信号通路在GIC的致癌作用中起关键作用。目前,针对GIC中Wnt/β-catenin信号传导的许多干预措施正在临床试验中进行测试,并获得了有希望的结果。不幸的是,目前尚无临床批准的有效靶向该途径的药物.本综述旨在评估靶向Wnt/β-catenin信号通路的临床治疗对GIC的影响。通过整合来自生物信息学数据库的数据和过去五年的最新文献,我们研究了GIC中Wnt/β-catenin通路基因和蛋白的异质性表达和调控机制。具体来说,我们专注于表达模式,突变频率,和临床预后,以了解它们对治疗策略的影响。此外,我们讨论了最近针对该途径的临床试验。了解目前正在进行临床研究的抑制剂可能有助于优化基础研究和临床策略。我们希望阐明针对Wnt/β-catenin通路的患者的精准治疗分层的现状将指导GIC的精准医学的未来创新。
    Gastrointestinal cancers (GICs) are highly prevalent cancers that threaten human health worldwide. The Wnt/β-catenin signaling pathway has been reported to play a pivotal role in the carcinogenesis of GICs. Numerous interventions targeting the Wnt/β-catenin signaling in GICs are currently being tested in clinical trials with promising results. Unfortunately, there are no clinically approved drugs that effectively target this pathway. This comprehensive review aims to evaluate the impact of clinical therapies targeting the Wnt/β-catenin signaling pathway in GICs. By integrating data from bioinformatics databases and recent literature from the past five years, we examine the heterogeneous expression and regulatory mechanisms of Wnt/β-catenin pathway genes and proteins in GICs. Specifically, we focus on expression patterns, mutation frequencies, and clinical prognoses to understand their implications for treatment strategies. Additionally, we discuss recent clinical trial efforts targeting this pathway. Understanding the inhibitors currently under clinical investigation may help optimize foundational research and clinical strategies. We hope that elucidating the current status of precision therapeutic stratification for patients targeting the Wnt/β-catenin pathway will guide future innovations in precision medicine for GICs.
    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
    目的:胃肠道肿瘤过表达电压门控钙(CaV3)通道(CaV3.1、3.2和3.3)。CaV3通道调节大肠癌细胞生长和凋亡。棉酚,一种在棉花植物中发现的多酚醛,具有抗肿瘤特性并抑制CaV3电流。对棉酚对CaV3通道的阻断机制及其在结肠癌细胞中的潜在抗癌作用进行了系统的研究,表达CaV3亚型。
    方法:使用公共存储库在胃肠道癌中和人类结直肠癌细胞系HCT116,SW480和SW620中分析了CaV3蛋白的转录物。通过结合异源表达系统和膜片钳实验研究了棉酚对CaV3通道的阻断机制。通过细胞增殖评估棉酚的抗肿瘤特性,活力和细胞周期测定。使用胞浆和内质网(ER)Ca2指标评估Ca2动力学。
    结果:高水平的CaV3转录物与胃肠道癌症的不良预后相关。棉酚对CaV3同工型的阻断是浓度和使用依赖性的相互作用,CaV3通道的激活和失活构象。棉酚和CaV3通道下调通过在G0/G1和G2/M期阻滞细胞周期抑制结直肠癌细胞增殖,分别。CaV3通道是结直肠癌细胞内质网对矢量Ca2吸收的基础。
    结论:棉酚差异阻断CaV3通道,其抗癌活性与大肠癌细胞中高水平的CaV3.1和CaV3.2相关。CaV3调节结直肠癌细胞的细胞增殖和Ca2+动力学。了解这种阻断机制可能会改善癌症治疗。
    OBJECTIVE: Gastrointestinal tumours overexpress voltage-gated calcium (CaV3) channels (CaV3.1, 3.2 and 3.3). CaV3 channels regulate cell growth and apoptosis colorectal cancer. Gossypol, a polyphenolic aldehyde found in the cotton plant, has anti-tumour properties and inhibits CaV3 currents. A systematic study was performed on gossypol blocking mechanism on CaV3 channels and its potential anticancer effects in colon cancer cells, which express CaV3 isoforms.
    METHODS: Transcripts for CaV3 proteins were analysed in gastrointestinal cancers using public repositories and in human colorectal cancer cell lines HCT116, SW480 and SW620. The gossypol blocking mechanism on CaV3 channels was investigated by combining heterologous expression systems and patch-clamp experiments. The anti-tumoural properties of gossypol were estimated by cell proliferation, viability and cell cycle assays. Ca2+ dynamics were evaluated with cytosolic and endoplasmic reticulum (ER) Ca2+ indicators.
    RESULTS: High levels of CaV3 transcripts correlate with poor prognosis in gastrointestinal cancers. Gossypol blockade of CaV3 isoforms is concentration- and use-dependent interacting with the closed, activated and inactivated conformations of CaV3 channels. Gossypol and CaV3 channels down-regulation inhibit colorectal cancer cell proliferation by arresting cell cycles at the G0/G1 and G2/M phases, respectively. CaV3 channels underlie the vectorial Ca2+ uptake by endoplasmic reticulum in colorectal cancer cells.
    CONCLUSIONS: Gossypol differentially blocked CaV3 channel and its anticancer activity was correlated with high levels of CaV3.1 and CaV3.2 in colorectal cancer cells. The CaV3 regulates cell proliferation and Ca2+ dynamics in colorectal cancer cells. Understanding this blocking mechanism maybe improve cancer therapies.
    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
    胃肠道癌症继续对全球健康构成重大挑战。每年诊断出数百万新病例。尽管在治疗方面取得了进展,许多患者的预后仍然较差。本文探讨了甘蓝酚的潜力,在各种藤黄物种中发现的聚异戊二烯化二苯甲酮,作为胃肠道恶性肿瘤的治疗剂。本文的目的是回顾最近对甘蓝酚抗癌特性的研究,其作用机制,和安全方面。Garcinol在食道中表现出抗癌作用,胃,结直肠,胰腺,和肝癌通过抑制转移,诱导细胞凋亡,靶向癌症进展中的关键分子途径。纳米技术被探索作为一种手段,以提高银花酚的递送和功效。安全性评估表明有希望的毒性特征。Garcinol显示出作为胃肠道癌症的天然治疗剂的巨大潜力,未来的研究需要优化它的交付,探索协同组合,并进行临床试验以验证其临床应用的有效性和安全性。
    Gastrointestinal cancers continue to pose a significant global health challenge, with millions of new cases diagnosed each year. Despite advancements in treatment, the prognosis for many patients remains poor. This article explores the potential of garcinol, a polyisoprenylated benzophenone found in various Garcinia species, as a therapeutic agent against gastrointestinal malignancies. The objective is to review recent research on garcinol\'s anticancer properties, its mechanisms of action, and safety aspects. Garcinol exhibits anticancer effects in esophageal, gastric, colorectal, pancreatic, and liver cancers by inhibiting metastasis, inducing apoptosis, and targeting key molecular pathways in cancer progression. Nanotechnology is explored as a means to enhance garcinol delivery and efficacy. Safety assessments suggest a promising toxicity profile. Garcinol shows significant potential as a natural therapeutic agent for gastrointestinal cancers, and future research is needed on optimizing its delivery, exploring synergistic combinations, and conducting clinical trials to validate its efficacy and safety for clinical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号