medicinal inorganic chemistry

药物无机化学
  • 文章类型: Journal Article
    尽管在过去的几十年中,癌症肿瘤的治疗有了显著的改善,癌症仍然是世界上最致命的疾病之一。为了克服目前应用的化疗药物的缺点,许多研究工作已经致力于铁凋亡诱导抗癌剂的开发。Ferroptosis是一种新描述的调节形式,非凋亡性细胞死亡与诊所内部的高潜力相关。在这里,据报道,Co(III)聚吡啶柳氮磺胺吡啶作为铁凋亡诱导剂的化学合成和生物学评估。进入癌细胞后,金属络合物主要积累在线粒体中,引发羟基自由基和脂质过氧化物的产生,最终通过铁凋亡导致细胞死亡。该化合物证明可以根除各种单层癌细胞以及结肠癌多细胞肿瘤球体。据我们所知,这项研究报告了能够诱导铁凋亡的Co(III)复合物的第一个实例。
    Despite significant improvements in the treatment of cancerous tumors in the last decades, cancer remains one of the deadliest diseases worldwide. To overcome the shortcomings of currently applied chemotherapeutic treatments, much research efforts have been devoted towards the development of ferroptosis inducing anticancer agents. Ferroptosis is a newly described form of regulated, non-apoptotic cell death that is associated with high potential inside the clinics. Herein, the chemical synthesis and biological evaluation of a Co(III) polypyridine sulfasalazine as a ferroptosis inducer is reported. Upon entering the cancerous cells, the metal complex primarily accumulated in the mitochondria, triggering the production of hydroxy radicals and lipid peroxides, ultimately causing cell death by ferroptosis. The compound demonstrated to eradicate various monolayer cancer cells as well as colon carcinoma multicellular tumor spheroids. To the best of our knowledge this study reports on the first example of a Co(III) complex that is capable of inducing ferroptosis.
    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
    当前的抗癌疗法存在诸如脱靶副作用和耐药性出现等问题;因此,发现替代治疗方法至关重要。这些可以包括开发具有不同作用方式的药物,以及新的生物分子靶标的探索。对于前者,人们对由外部刺激激活以产生细胞毒性物质的药物越来越感兴趣。对于后者,正在努力探索非规范的DNA和RNA结构(例如鸟嘌呤-四链体),作为替代的生物分子靶标。在这里,我们报告了21个新的铂(II)-Salphen络合物库的合成,研究它们的光物理和光化学性质,它们与双链体和四链体DNA的相互作用,以及它们在黑暗和光照下对HeLa癌细胞的细胞毒性。由于铂(II)配合物的固有磷光,共聚焦显微镜用于六种复合物,以确定其细胞通透性和在两种癌细胞系中的定位。这些研究使我们能够鉴定出两种具有高鸟嘌呤-四链体DNA亲和力和选择性的铅铂(II)复合物,良好的细胞通透性和核定位,照射后对HeLa癌细胞具有高细胞毒性,在黑暗中未检测到细胞毒性。
    Current anticancer therapies suffer from issues such as off-target side effects and the emergence of drug resistance; therefore, the discovery of alternative therapeutic approaches is vital. These can include the development of drugs with different modes of action, and the exploration of new biomolecular targets. For the former, there has been increasing interest in drugs that are activated by an external stimulus to generate cytotoxic species. For the latter, significant efforts are being directed to explore non-canonical DNA and RNA structures (e.g. guanine-quadruplexes), as alternative biomolecular targets. Herein we report the synthesis of a library of 21 new platinum(II)-Salphen complexes, investigation of their photophysical and photochemical properties, their interactions with duplex and quadruplex DNA, and their cytotoxicity against HeLa cancer cells in the dark and upon light irradiation. Thanks to the intrinsic phosphorescence of the platinum(II) complexes, confocal microscopy was used for six of the complexes to determine their cellular permeability and localisation in two cancer cell lines. These studies have allowed us to identify two lead platinum(II) complexes with high guanine-quadruplex DNA affinity and selectivity, good cell permeability and nuclear localisation, and high cytotoxicity against HeLa cancer cells upon irradiation with no detected cytotoxicity in the dark.
    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
    肿瘤内Pt(IV)前药的选择性激活已成为肿瘤治疗中的有希望的策略。尽管光激活和超声激活的Pt(IV)前药已取得进展,对于光敏剂(PS)的非特异性激活以及光毒性和化学毒性的可能性仍然存在担忧。在这项研究中,开发了一种可以被酸性肿瘤微环境和光激活的连续双锁铂(IV)纳米前药。通过将PS锁定的Pt(IV)与聚合物核缀合来制备Pt(IV)前药,然后与金属铁螯合以锁定其光活性并形成金属纳米前药。在酸性肿瘤微环境条件下,金属纳米前药经历铁的解离,在光照下引发奥沙利铂的还原过程,导致化疗和光动力疗法(PDT)的激活。此外,前药可以诱导金属触发的铁凋亡和肿瘤相关巨噬细胞(TAM)的极化,从而增强肿瘤抑制。在纳米颗粒递送系统中采用的双锁策略代表了铂基抗癌药物应用的扩展。使其成为癌症治疗的新方向。
    Selective activation of Pt(IV) prodrugs within tumors has emerged as a promising strategy in tumor treatment. Although progress has been made with photo- and ultrasound-activated Pt(IV) prodrugs, concerns remain over the non-specific activation of photosensitizers (PS) and the potential for phototoxicity and chemical toxicity. In this study, a sequential dual-locked Pt(IV) nano-prodrug that can be activated by both the acidic tumor microenvironment and light was developed. The Pt(IV) prodrug was prepared by conjugating PS-locked Pt(IV) to a polymeric core, which was then chelated with metallo iron to lock its photoactivity and form a metallo-nano prodrug. Under acidic tumor microenvironment conditions, the metallo-nano prodrug undergoes dissociation of iron, triggering a reduction process in oxaliplatin under light irradiation, resulting in the activation of both chemotherapy and photodynamic therapy (PDT). Additionally, the prodrug could induce metallo-triggered ferroptosis and polarization of tumor-associated macrophages (TAM), thereby enhancing tumor inhibition. The dual-lock strategy employed in a nanoparticle delivery system represents an expansion in the application of platinum-based anticancer drugs, making it a promising new direction in cancer 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
    癌症是全世界最致命的疾病之一。对抗这种疾病最常用的治疗技术之一是化疗。尽管取得了成功,大多数临床应用的化疗药物都有很强的副作用和耐药性。为了克服这个限制,许多研究工作都致力于开发新的抗癌剂。在最有前途的一类化合物中,Cu(Ⅱ)配合物涌现。尽管它们有很强的细胞毒作用,这些药物通常与低水溶性有关,稳定性低,肿瘤选择性差。为了克服这些限制,在这里,我们报道了将有希望的Cu(II)三吡啶络合物与PluronicF-127/泊洛沙姆-407聚合物载体封装到纳米颗粒中。除了克服药理学上的缺点,纳米颗粒能够根除人乳腺癌单层细胞,并在纳摩尔浓度下挑战多细胞肿瘤球体。
    Cancer is one of the deadliest diseases worldwide. One of the most commonly applied therapeutic techniques to combat this disease is chemotherapy. Despite its success, the majority of clinically applied chemotherapeutic agents are associated with strong side effects and drug resistance. To overcome this limitation, much research efforts are devoted toward the development of new anticancer agents. Among the most promising class of compounds, Cu(II) complexes have emerged. Despite their strong cytotoxic effect, these agents are typically associated with low water solubility, low stability, and poor tumor selectivity. To overcome these limitations, herein, we report on the encapsulation of a promising Cu(II) terpyridine complex with the Pluronic F-127/Poloxamer-407 polymeric carrier into nanoparticles. Besides overcoming the pharmacological drawbacks, the nanoparticles were able to eradicate human breast adenocarcinoma monolayer cells as well as challenging multicellular tumor spheroids at nanomolar concentrations.
    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
    最佳的癌症化疗方案应有效解决肿瘤的耐药性,同时引发抗肿瘤免疫反应。研究表明,非凋亡性细胞死亡,比如焦化凋亡和铁化凋亡,能增强免疫反应。尽管如此,关于肿瘤发病机制及其与免疫反应的相关性的研究和报道有限。在这里,我们设计并合成了靶向细胞核和线粒体的Ru(II)复合物,以诱导细胞致癌。简而言之,Ru(II)复合物破坏细胞核和线粒体DNA,其中有活性的多聚ADP-核糖聚合酶1,伴随着ATP消耗和波利明激活。同时,线粒体损伤和内质网应激导致Ca2+离子的释放和Calpain1的表达增加。随后,特定的孔蛋白porimin和Calpain1促进cr破坏或空泡化,最终导致细胞膜破裂.RNA测序的分析表明,Ru(II)复合物可以启动与致癌相关的途径并激活先天和适应性免疫。体内实验已证实,致癌促进树突状细胞的成熟和适应性细胞毒性T淋巴细胞的唤醒,但也诱导肿瘤相关巨噬细胞(TAM)向M1表型的极化,并激活TAM的先天免疫反应。
    An optimal cancer chemotherapy regimen should effectively address the drug resistance of tumors while eliciting antitumor-immune responses. Research has shown that non-apoptotic cell death, such as pyroptosis and ferroptosis, can enhance the immune response. Despite this, there has been limited investigation and reporting on the mechanisms of oncosis and its correlation with immune response. Herein, we designed and synthesized a Ru(II) complex that targeted the nucleus and mitochondria to induce cell oncosis. Briefly, the Ru(II) complex disrupts the nucleus and mitochondria DNA, which active polyADP-ribose polymerase 1, accompanied by ATP consumption and porimin activation. Concurrently, mitochondrial damage and endoplasmic reticulum stress result in the release of Ca2+ ions and increased expression of Calpain 1. Subsequently, specific pore proteins porimin and Calpain 1 promote cristae destruction or vacuolation, ultimately leading to cell membrane rupture. The analysis of RNA sequencing demonstrates that the Ru(II) complex can initiate the oncosis-associated pathway and activate both innate and adaptive immunity. In vivo experiments have confirmed that oncosis promotes dendritic cell maturation and awakens adaptive cytotoxic T lymphocytes but also activates the innate immune by inducing the polarization of macrophages towards an M1 phenotype.
    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
    肿瘤转移和复发被认为是癌症相关死亡的主要原因。作为一种新兴的治疗方法,越来越多的研究努力致力于免疫原性细胞死亡(ICD)诱导化合物,以解决这一挑战。临床上批准的化学治疗性Pt络合物不能或仅仅能很差地触发ICD。在这里,据报道,具有全氟化碳链的Pt(II)络合物顺铂的轴向功能化为ICD诱导的Pt(IV)前药。引人注目的是,虽然Pt(II)配合物以及全氟化碳配体没有诱导ICD,Pt(IV)前药意外地证明了通过内质网中的积累和该细胞器中活性氧的产生来诱导ICD。为了增强药理特性,将该化合物与人血清白蛋白封装在纳米颗粒中。当选择性地在肿瘤组织中积累时,纳米颗粒在小鼠模型中对骨肉瘤具有很强的肿瘤生长抑制作用。体内肿瘤疫苗分析也证明了Pt(IV)是理想的ICD诱导物的能力。总的来说,这项研究报道了轴向全氟化碳链修饰的Pt(IV)复合物用于骨肉瘤的ICD诱导和化学免疫疗法。
    Tumor metastases and reoccurrence are considered the leading causes of cancer-associated deaths. As an emerging therapeutic method, increasing research efforts have been devoted to immunogenic cell death (ICD)-inducing compounds to solve the challenge. The clinically approved chemotherapeutic Pt complexes are not or are only poorly able to trigger ICD. Herein, the axial functionalization of the Pt(II) complex cisplatin with perfluorocarbon chains into ICD-inducing Pt(IV) prodrugs is reported. Strikingly, while the Pt(II) complex as well as the perfluorocarbon ligands did not induce ICD, the Pt(IV) prodrug demonstrated unexpectantly the induction of ICD through accumulation in the endoplasmic reticulum and generation of reactive oxygen species in this organelle. To enhance the pharmacological properties, the compound was encapsulated with human serum albumin into nanoparticles. While selectively accumulating in the tumorous tissue, the nanoparticles demonstrated a strong tumor growth inhibitory effect against osteosarcoma inside a mouse model. In vivo tumor vaccine analysis also demonstrated the ability of Pt(IV) to be an ideal ICD inducer. Overall, this study reports on axially perfluorocarbon chain-modified Pt(IV) complexes for ICD induction and chemoimmunotherapy in osteosarcoma.
    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
    癌症是全世界最致命的疾病之一。化疗仍然是抗癌治疗的最主要形式之一。尽管他们在临床上取得了成功,使用的化学治疗剂与严重的副作用和药理学限制有关。为了克服这些缺点,需要开发新类型的化学治疗剂。在这里,提出了作为潜在化疗药物候选物的双核铼(I)配合物的化学合成和生物学评估。发现金属络合物通过能量依赖性胞吞途径内化,初级积累在线粒体中。铼(I)复合物证明通过凋亡诱导针对微摩尔范围内的多种癌细胞的细胞死亡。先导化合物显示以微摩尔浓度根除胰腺癌多细胞肿瘤球体。
    Cancer is one of the deadliest diseases worldwide. Chemotherapy remains one of the most dominant forms for anticancer treatment. Despite their clinical success, the used chemotherapeutic agents are associated with severe side effect and pharmacological limitations. To overcome these drawbacks there is a need for the development of new types of chemotherapeutic agents. Herein, the chemical synthesis and biological evaluation of dinuclear rhenium(I) complexes as potential chemotherapeutic drug candidates are proposed. The metal complexes were found to be internalized by an energy dependent endocytosis pathway, primary accumulating in the mitochondria. The rhenium(I) complexes demonstrated to induce cell death against a variety of cancer cells in the micromolar range through apoptosis. The lead compound showed to eradicate a pancreatic carcinoma multicellular tumor spheroid at micromolar concentrations.
    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
    自从最后一种螯合剂在临床上被批准用于对抗有毒金属以来,它将很快20年。尽管金属中毒已经为人类所知了几个世纪,只有大约十几种化合物,所有这些都是小分子,组成药物工具箱,以驱逐本质上有毒或必需但监管不当的金属。这些化合物广泛地具有各种缺点,最关键的是,金属选择性差。药物无机化学能否为这些旧挑战提供现代解决方案?从这个角度来看,描述了利用短肽进行螯合治疗的机会和优势。虽然广泛旨在解决各种有毒金属,用肽靶向铅(Pb)的成就揭示了隐藏在该化学空间中的未开发潜力,并提高了肽可能改革螯合疗法的可能性。
    It will soon be twenty years since the last chelating agent was clinically approved to be used against toxic metals. Even though metal poisoning has been known to humankind for centuries, only about a dozen compounds, all of which are small molecules, compose the pharmaceutical toolbox to expel intrinsically toxic or essential but misregulated metals. These compounds widely suffer from various drawbacks, most critically, poor metal selectivity. Can medicinal inorganic chemistry offer modern solutions to these old challenges? In this perspective, the opportunities and advantages of harnessing short peptides for chelation therapy are described. While broadly aiming to address various toxic metals, achievements in targeting lead (Pb) with peptides reveal the unexplored potential hidden in this chemical space and raise the possibility that peptides may reform chelation 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
    化疗仍然是术后骨肉瘤的主要治疗方法。然而,骨肉瘤化疗缺乏选择性导致不可预测的治疗效果,不良副作用和耐药性。设计与阿仑膦酸盐(ALN)和脂质尾部共价结合的铂(IV)(PtIV)前药两亲物以克服该限制。ALN-PtIV-Lipo可以自组装成PtIV脂质纳米粒(APtIV),用于骨肉瘤靶向化疗和骨破坏抑制。我们证明,与顺铂相比,APtIV在根除骨肉瘤细胞方面实现了8倍的增加,并且在体外通过APtIV对骨肉瘤细胞的选择性抑制是乳腺癌细胞的3倍。静脉注射后,APtIV在体内有效积累在骨肉瘤部位,导致原发性骨肉瘤生长显著抑制(TGI=86%),和减轻骨骼破坏。因此,APtIV为骨肉瘤中增强化疗靶向和骨破坏抑制提供了有希望的解决方案。本文受版权保护。保留所有权利。
    Chemotherapy remains the primary treatment method for osteosarcoma after surgery. However, the lack of selectivity of chemotherapy for osteosarcoma leads to unpredictable therapeutic effects, undesirable side effects, and drug resistance. A platinum(IV) (PtIV ) prodrug amphiphile (ALN-PtIV -Lipo) covalently bound to alendronate (ALN) and a lipid tail is designed to overcome these limitations. ALN-PtIV -Lipo can self-assemble into PtIV lipid nanoparticles (APtIV ) for osteosarcoma targeting chemotherapy and bone destruction inhibition. It is demonstrated that APtIV achieved an eightfold increase in the eradication of osteosarcoma cells compared to cisplatin and threefold selective inhibition of osteosarcoma cells over breast cancer cells via APtIV in vitro. After intravenous injection, APtIV effectively accumulates at the osteosarcoma site in vivo, resulting in significantly suppressed primary osteosarcoma growth, and alleviation of bone destruction. Therefore, APtIV delivers a promising solution for enhanced chemotherapy targeting and bone destruction inhibition in osteosarcoma.
    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
    动力学不稳定性或反应性对体外细胞毒性的影响,在等离子体中的稳定性,体内肿瘤和组织积聚,和含有OOβ-二酮酸盐离开配体的铂(II)(Pt)抗癌剂的抗肿瘤功效仍未被探索。为了调查这一点,我们合成了Pt配合物[(NH3)2Pt(L1-H)]NO3和[(DACH)Pt(L1-H)]NO3(L1=4,4,4-三氟-1-二茂铁基丁烷-1,3-二酮,DACH=1R,2R-环己烷-1,2-二胺),含有缺电子的L1-HOO离开配体和[(NH3)2Pt(L2-H)]NO3和[(DACH)Pt(L2-H)]NO3(L2=1-二茂铁基丁烷-1,3-二酮),其中含有富电子的L2-HOO离开。虽然所有四种复合物都具有相当的亲脂性,发现吸电子CF3基团的存在显着增强了这些复合物对亲核生物分子的反应性。体外细胞测定显示,与其较低反应性的类似物相比,更具反应性的复合物具有更高的细胞摄取和更高的抗癌效力。但在体内情况正好相反,与活性较高的类似物相比,活性较低的复合物在带有卵巢异种移植的小鼠中显示出改善的组织和肿瘤积累以及更好的抗癌功效。最后,除了证明动力学不稳定性对体外和体内抗肿瘤效力的深刻但对比的影响,我们还描述了动力学不稳定性对这类配合物作用机理的影响。
    The impact of kinetic lability or reactivity on in vitro cytotoxicity, stability in plasma, in vivo tumor and tissue accumulation, and antitumor efficacy of functional platinum(II) (Pt) anticancer agents containing a O˄O β-diketonate leaving ligand remain largely unexplored. To investigate this, we synthesized Pt complexes [(NH3 )2 Pt(L1-H)]NO3 and [(DACH)Pt(L1-H)]NO3 (L1=4,4,4-trifluoro-1-ferrocenylbutane-1,3-dione, DACH=1R,2R-cyclohexane-1,2-diamine) containing an electron deficient [L1-H]- O˄O leaving ligand and [(NH3 )2 Pt(L2-H)]NO3 and [(DACH)Pt(L2-H)]NO3 (L2=1-ferrocenylbutane-1,3-dione) containing an electron-rich [L2-H]- O˄O leaving ligand. While all four complexes have comparable lipophilicity, the presence of the electron-withdrawing CF3 group was found to dramatically enhance the reactivity of these complexes toward nucleophilic biomolecules. In vitro cellular assays revealed that the more reactive complexes have higher cellular uptake and higher anticancer potency as compared to their less reactive analogs. But the scenario is opposite in vivo, where the less reactive complex showed improved tissue and tumor accumulation and better anticancer efficacy in mice bearing ovarian xenograft when compared to its more reactive analog. Finally, in addition to demonstrating the profound but contrasting impact of kinetic lability on in vitro and in vivo antitumor potencies, we also described the impact of kinetic lability on the mechanism of action of this class of promising antitumor agents.
    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