Glutaminase inhibitors

  • 文章类型: Journal Article
    癌症对人类健康和生命构成重大威胁。化疗,免疫疗法和化学动力学疗法(CDT)是癌症的有效治疗方法。然而,肿瘤细胞中通过谷氨酰胺进行代谢重编程的存在限制了其治疗效果.在这里,我们提出了一种有效的组装策略来合成一种新型的基于金属-多酚的多功能纳米药物(Fe-DBEF),其中包含PluronicF127稳定的铁离子交联的表没食子儿茶素没食子酸酯(EGCG)纳米颗粒,负载有GLS1抑制剂双-2-(5-苯基乙酰氨基-1,3,4-噻二唑-2-基)乙基硫醚(BPTES)和我们的研究表明,通过结合体外细胞实验,Fe-DBEF纳米药物在胰腺癌中表现出高效的抗增殖特性,人体类器官实验和KPC动物实验。值得注意的是,Fe-DBEF纳米药物可以减少肿瘤细胞中谷胱甘肽(GSH)的产生,从而降低其对ROS治疗的抗性。此外,过量的ROS产生也会加剧DOX引起的DNA损伤,协同增敏化疗和促进细胞凋亡有效治疗胰腺癌。总的来说,我们的研究结果表明,使用基于金属-多酚的多功能纳米药物抑制谷氨酰胺代谢以增加化疗/CDT的敏感性,为胰腺癌的治疗提供了一种有前景的多种治疗手段的组合.
    Cancer poses a significant threat to human health and life. Chemotherapy, immunotherapy and chemodynamic therapy (CDT) are effective treatments for cancer. However, the presence of metabolic reprogramming via glutamine in tumor cells limits their therapeutic effectiveness. Herein, we propose an effective assembly strategy to synthesize a novel metal-polyphenolic based multifunctional nanomedicine (Fe-DBEF) containing Pluronic F127 stable ferric ion crosslinked epigallocatechin gallate (EGCG) nanoparticles loaded with GLS1 inhibitor bis-2-(5-phenylacetamino-1,3,4-thiadiazole-2-yl) ethyl sulfide (BPTES) and chemotherapy drug doxorubicin (DOX). Our study demonstrates that Fe-DBEF nanomedicine exhibits high efficiency anti-proliferation properties in pancreatic cancer through a combination of in vitro cell experiments, human organoid experiments and KPC animal experiments. Notably, Fe-DBEF nanomedicine can reduce the production of glutathione (GSH) in tumor cells, thereby reducing their resistance to ROS therapy. Additionally, excessive ROS production also aggravates DNA damage caused by DOX, synergistically sensitizing chemotherapy and promoting apoptosis for efficient treatment of pancreatic cancer. Overall, our findings suggest that inhibiting glutamine metabolism to increase the sensitivity of chemotherapy/CDT using metal-polyphenolic based multifunctional nanomedicine provides a promising combination of multiple therapeutic means for treating pancreatic 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
    癌症代谢现在是治疗干预的关键领域,靶向独特的代谢重编程对于肿瘤生长和存活至关重要。本文回顾了通过糖酵解和谷氨酰胺酶抑制剂解决代谢脆弱性的治疗潜力,破坏癌细胞的新陈代谢.讨论了肿瘤异质性和适应性抗性等挑战,采用包括个性化医疗和预测生物标志物在内的策略来提高治疗效果。此外,将饮食和生活方式的改变与代谢靶向相结合,强调了改善治疗结果的整体方法.本文还研究了将这些策略纳入标准护理的好处,强调更有可能量身定制,更安全的治疗方法总之,利用代谢漏洞预示着肿瘤学的新时代,将代谢靶向定位在个性化癌症治疗和转变患者护理的最前沿。
    Cancer metabolism is now a key area for therapeutic intervention, targeting unique metabolic reprogramming crucial for tumor growth and survival. This article reviews the therapeutic potential of addressing metabolic vulnerabilities through glycolysis and glutaminase inhibitors, which disrupt cancer cell metabolism. Challenges such as tumor heterogeneity and adaptive resistance are discussed, with strategies including personalized medicine and predictive biomarkers to enhance treatment efficacy. Additionally, integrating diet and lifestyle changes with metabolic targeting underscores a holistic approach to improving therapy outcomes. The article also examines the benefits of incorporating these strategies into standard care, highlighting the potential for more tailored, safer treatments. In conclusion, exploiting metabolic vulnerabilities promises a new era in oncology, positioning metabolic targeting at the forefront of personalized cancer therapy and transforming patient care.
    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
    靶向治疗已经彻底改变了肾细胞癌(RCC)的治疗。VHL/HIF途径负责氧稳态的调节并且在RCC中经常改变。靶向该途径以及mTOR途径在治疗RCC方面取得了显著进展。这里,我们回顾了治疗RCC最有前途的新型靶向疗法,包括HIF2α,MET,代谢靶向,和表观基因组重编程。
    Targeted therapies have revolutionized the treatment of renal cell carcinoma (RCC). The VHL/HIF pathway is responsible for the regulation of oxygen homeostasis and is frequently altered in RCC. Targeting this pathway as well as the mTOR pathway have yielded remarkable advances in the treatment of RCC. Here, we review the most promising novel targeted therapies for the treatment of RCC, including HIF2α, MET, metabolic targeting, and epigenomic reprogramming.
    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
    谷氨酰胺代谢是几种癌症的重要标志,在胶质母细胞瘤癌细胞(GBM)中具有证明的抗肿瘤活性。GBM细胞通过谷氨酰胺分解过程调节谷氨酰胺并将其用作其增殖的主要能量来源。酶,例如谷氨酰胺分解中的谷氨酰胺酶,可以被小分子抑制剂靶向,从而显示出有希望的抗癌特性。对谷氨酰胺分解的抗性需要开发新的治疗分子来克服耐药性。在这里,我们已经报道了一个新的限制甲基苯并[b,f][1,5]二氧辛衍生物作为谷氨酰胺酶(GLS)抑制剂及其抗GBM潜力。通过2'-羟基苯乙酮的自缩合获得由七个分子组成的文库,其中三个分子,即化合物3、5和6用GLS(PDBID;4O7D)鉴定为具有范围在-10.2和-9.8kcal/mol之间的更高的结合能值。这些化合物的药理学验证在GBM细胞中也显示出比标准药物替莫唑胺(TMZ)更高的生长抑制作用。最有前途的化合物,6,遵守Lipinski的5条规则,并被鉴定与GLS的关键残基Arg307,Asp326,Lys328,Lys399和Glu403相互作用。该化合物在LN229和SNB19细胞中表现出最佳的细胞毒性作用,IC50值为63μM和83μM,分别。最佳约束二苯并[b,f][1,5]dioxocininthetestedseriesincreasedprotocoatingviareactiveoxygenspeciesproductioninbothGBMcells,并随着时间的推移在两种细胞系中均表现出抗迁移和抗增殖特性。我们的结果强调了二苯并[b,f][1,5]dioxocin从结构的基础上,并证明谷氨酰胺分解的抑制可能有助于GBM治疗的药理学干预。
    Glutamine metabolism is an important hallmark of several cancers with demonstrated antitumor activity in glioblastoma cancer cells (GBM). GBM cells regulate glutamine and use it as a major energy source for their proliferation through the glutaminolysis process. Enzymes, such as glutaminase in glutaminolysis, can be targeted by small-molecule inhibitors, thus exhibiting promising anticancer properties. The resistance to glutaminolysis demands the development of new therapeutic molecules to overcome drug resistance. Herein, we have reported a novel library of constrained methanodibenzo[b,f][1,5]dioxocin derivatives as glutaminase (GLS) inhibitors and their anti-GBM potential. The library consisting of seven molecules was obtained through self-condensation of 2\'-hydroxyacetophenones, out of which three molecules, namely compounds 3, 5, and 6, were identified with higher binding energy values ranging between -10.2 and -9.8 kcal/mol with GLS (PDB ID; 4O7D). Pharmacological validation of these compounds also showed a higher growth inhibition effect in GBM cells than the standard drug temozolomide (TMZ). The most promising compound, 6, obeyed Lipinski\'s rule of five and was identified to interact with key residues Arg307, Asp326, Lys328, Lys399, and Glu403 of GLS. This compound exhibited the best cytotoxic effect with IC50 values of 63 µM and 83 µM in LN229 and SNB19 cells, respectively. The potential activation of GLS by the best-constrained dibenzo[b,f][1,5]dioxocin in the tested series increased apoptosis via reactive oxygen species production in both GBM cells, and exhibited anti-migratory and anti-proliferative properties over time in both cell lines. Our results highlight the activation mechanism of a dibenzo[b,f][1,5]dioxocin from the structural basis and demonstrate that inhibition of glutaminolysis may facilitate the pharmacological intervention for GBM 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
    乳腺癌(BC)是一种异质性癌症,具有多种亚型,影响全球女性。三阴性乳腺癌(TNBC)是一种突出的BC亚型,预后差,具有侵袭性表型。最近对代谢重编程的理解支持其在癌细胞生长及其对其微环境的适应中的作用。Warburg效应的特征在于从氧化代谢转变为还原代谢和乳酸的外部分泌。Warburg效应阻止了通过丙酮酸脱氢酶失活而在三羧酸(TCA)循环中使用所需的丙酮酸。因此,它是促进糖酵解和破坏TCA循环的主要调节机制。谷氨酰胺(Gln)可以为癌细胞提供补充能量。此外,它是支持癌细胞中生物能量学和生物合成活性的主要底物,并在一系列其他过程中发挥着至关重要的作用,例如铁凋亡。因此,在TCA循环中,葡萄糖向还原性Gln代谢的转换是通过Warburg效应进行的缺氧诱导因子(HIF)进行的。文献表明,TNBC对Gln的成瘾可以促进这些癌症的增殖和侵袭性。因此,Gln代谢抑制剂,如CB-839,可用于管理TNBC的致癌特性。这种抑制剂,以及传统的化疗药物,可以潜在地提高TNBC治疗的效率和疗效。在这次审查中,我们从Gln代谢抑制剂可以改善当前化疗药物作用的角度讨论了葡萄糖和Gln代谢与癌细胞生长控制之间的关系.
    Breast cancer (BC) is a heterogeneous cancer with multiple subtypes affecting women worldwide. Triple-negative breast cancer (TNBC) is a prominent subtype of BC with poor prognosis and an aggressive phenotype. Recent understanding of metabolic reprogramming supports its role in the growth of cancer cells and their adaptation to their microenvironment. The Warburg effect is characterized by the shift from oxidative to reductive metabolism and external secretion of lactate. The Warburg effect prevents the use of the required pyruvate in the tricarboxylic acid (TCA) cycle progressing through pyruvate dehydrogenase inactivation. Therefore, it is a major regulatory mechanism to promote glycolysis and disrupt the TCA cycle. Glutamine (Gln) can supply the complementary energy for cancer cells. Additionally, it is the main substrate to support bioenergetics and biosynthetic activities in cancer cells and plays a vital role in a wide array of other processes such as ferroptosis. Thus, the switching of glucose to Gln in the TCA cycle toward reductive Gln metabolism is carried out by hypoxia-inducible factors (HIFs) conducted through the Warburg effect. The literature suggests that the addiction of TNBC to Gln could facilitate the proliferation and invasiveness of these cancers. Thus, Gln metabolism inhibitors, such as CB-839, could be applied to manage the carcinogenic properties of TNBC. Such inhibitors, along with conventional chemotherapy agents, can potentially improve the efficiency and efficacy of TNBC treatment. In this review, we discuss the associations between glucose and Gln metabolism and control of cancer cell growth from the perspective that Gln metabolism inhibitors could improve the current chemotherapy drug effects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Metabolic reprogramming in cancer targets glutamine metabolism as a key mechanism to provide energy, biosynthetic precursors and redox requirements to allow the massive proliferation of tumor cells. Glutamine is also a signaling molecule involved in essential pathways regulated by oncogenes and tumor suppressor factors. Glutaminase isoenzymes are critical proteins to control glutaminolysis, a key metabolic pathway for cell proliferation and survival that directs neoplasms\' fate. Adaptive glutamine metabolism can be altered by different metabolic therapies, including the use of specific allosteric inhibitors of glutaminase that can evoke synergistic effects for the therapy of cancer patients. We also review other clinical applications of in vivo assessment of glutaminolysis by metabolomic approaches, including diagnosis and monitoring of cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    BACKGROUND: Metabolic reprogramming of tumours is a hallmark of cancer. Among the changes in the metabolic network of cancer cells, glutaminolysis is a key reaction altered in neoplasms. Glutaminase proteins control the first step in glutamine metabolism and their expression correlates with malignancy and growth rate of a great variety of cancers. The two types of glutaminase isoenzymes, GLS and GLS2, differ in their expression patterns and functional roles: GLS has oncogenic properties and GLS2 has been described as a tumour suppressor factor.
    RESULTS: We have focused on glutaminase connections with key oncogenes and tumour suppressor genes. Targeting glutaminase isoenzymes includes different strategies aimed at deactivating the rewiring of cancer metabolism. In addition, we found a long list of metabolic enzymes, transcription factors and signalling pathways dealing with glutaminase. On the other hand, a number of chemicals have been described as isoenzyme-specific inhibitors of GLS and/or GLS2 isoforms. These molecules are being characterized as synergic and therapeutic agents in many types of tumours.
    CONCLUSIONS: This review states the metabolic pathways that are rewired in cancer, the roles of glutaminase isoforms in cancer, as well as the metabolic circuits regulated by glutaminases. We also show the plethora of anticancer drugs that specifically inhibit glutaminase isoenzymes for treating several sets of 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
    Altered cellular metabolism is a hallmark of cancer. Cancer cells express isoforms of metabolic enzymes that may constitute therapeutic targets. Glutaminase controls glutamine metabolism and their expression correlate with malignancy of tumours. The two types of glutaminase isoenzymes, GLS and GLS2, differ in their expression patterns and functional roles: GLS has oncogenic properties and GLS2 has been described as a tumour suppressor factor. Selective genomic and epigenomic intervention over glutaminase affects the metabolic reprogramming of cancer. This review highlights the molecular metabolic vulnerabilities in various types of cancer, to be used for biomarker development, drug design, and in personalized oncology.
    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
    Metabolomics, the newest of the omics sciences that also include genomics, transcriptomics and proteomics, has matured into a reliable high-throughput technology. Gas chromatography combined with time-of-flight mass spectrometry (GC-TOFMS) is a suitable method to analyze the central metabolism in fresh frozen tumor tissue samples. Bioinformatics methods, including the PROFILE clustering developed by us, permit integrated analysis and fast interpretation of metabolomics data in the context of enzymatic reactions and metabolic pathways. The metabolome analyses of three solid tumor types presented here, together with the results of other authors, show that metabolites are suitable as biomarkers and provide diverse options for translation into the clinical setting.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    卵巢癌的特征是细胞能量代谢增加,主要由葡萄糖和谷氨酰胺满足。靶向代谢途径是提高卵巢癌治疗效果和潜在克服耐药性的有吸引力的方法。在铂敏感的卵巢癌细胞系中,两者的代谢,葡萄糖和谷氨酰胺最初是响应铂治疗而上调的。相比之下,铂抗性细胞显示出对谷氨酰胺存在的显著依赖性,谷氨酰胺转运体ASCT2和谷氨酰胺酶的表达上调。与铂敏感细胞系相比,这导致更高的耗氧率,这反映了通过三羧酸循环对谷氨酰胺利用的依赖性增加。谷氨酰胺酶的稳定过表达证实了谷氨酰胺代谢的重要作用,这赋予了铂电阻。相反,在铂抗性细胞中谷氨酰胺酶的shRNA敲除导致对铂处理的再敏化。重要的是,谷氨酰胺酶抑制剂BPTES与铂在体外协同抑制铂敏感和耐药卵巢癌。与单独的任一处理相比,使用铂与BPTES一起显著增加了凋亡诱导。我们的发现表明,靶向谷氨酰胺代谢与基于铂的化疗一起提供了一种潜在的治疗策略,特别是在耐药卵巢癌中。
    Ovarian cancer is characterized by an increase in cellular energy metabolism, which is predominantly satisfied by glucose and glutamine. Targeting metabolic pathways is an attractive approach to enhance the therapeutic effectiveness and to potentially overcome drug resistance in ovarian cancer. In platinum-sensitive ovarian cancer cell lines the metabolism of both, glucose and glutamine was initially up-regulated in response to platinum treatment. In contrast, platinum-resistant cells revealed a significant dependency on the presence of glutamine, with an upregulated expression of glutamine transporter ASCT2 and glutaminase. This resulted in a higher oxygen consumption rate compared to platinum-sensitive cell lines reflecting the increased dependency of glutamine utilization through the tricarboxylic acid cycle. The important role of glutamine metabolism was confirmed by stable overexpression of glutaminase, which conferred platinum resistance. Conversely, shRNA knockdown of glutaminase in platinum resistant cells resulted in re-sensitization to platinum treatment. Importantly, combining the glutaminase inhibitor BPTES with platinum synergistically inhibited platinum sensitive and resistant ovarian cancers in vitro. Apoptotic induction was significantly increased using platinum together with BPTES compared to either treatment alone. Our findings suggest that targeting glutamine metabolism together with platinum based chemotherapy offers a potential treatment strategy particularly in drug resistant ovarian cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号