synergistic therapy

协同治疗
  • 文章类型: Journal Article
    胶质母细胞瘤(GBM)是最常见和侵袭性的恶性脑肿瘤。标准治疗包括最大限度的手术切除,放射治疗,和佐剂替莫唑胺(TMZ)给药。然而,TMZ抗性的快速发展和血脑屏障(BBB)的不通透性显着阻碍了治疗效果。在这里,我们开发了载有TMZ和氯硝柳胺(NIC)的时空控制微针贴片(BMN)以克服GBM抗性。我们发现透明质酸(HA)增加了牛血清白蛋白(BSA)的粘度,并证明BSA/HA的浓度对暴露于高温处理的降解率产生影响。表明较高的BSA/HA浓度导致较慢的药物释放。为了优化药物释放速率并确保协同抗肿瘤作用,选择构成BMN底部的15%BSA/HA溶液来装载TMZ,显示药物持续释放超过28天,保证TMZ抗性细胞(U251-TR)的长期DNA损伤。由装载有NIC的10%BSA/HA溶液制成的针尖在14天内释放药物,通过抑制O6-甲基鸟嘌呤-DNA甲基转移酶(MGMT)的活性来增强TMZ的功效。BMN表现出优异的机械性能,绕过BBB,并逐渐将药物释放到肿瘤周围,从而显著抑制肿瘤增殖和扩大小鼠中位生存期。BMNs补丁的按需交付显示出强大的临床应用转化潜力,特别是在协同GBM治疗中。
    Glioblastoma (GBM) is the most common and aggressive malignant brain tumor. Standard therapy includes maximal surgical resection, radiotherapy, and adjuvant temozolomide (TMZ) administration. However, the rapid development of TMZ resistance and the impermeability of the blood-brain barrier (BBB) significantly hinder the therapeutic efficacy. Herein, we developed spatiotemporally controlled microneedle patches (BMNs) loaded with TMZ and niclosamide (NIC) to overcome GBM resistance. We found that hyaluronic acid (HA) increased the viscosity of bovine serum albumin (BSA) and evidenced that concentrations of BSA/HA exert an impact degradation rates exposure to high-temperature treatment, showing that the higher BSA/HA concentrations result in slower drug release. To optimize drug release rates and ensure synergistic antitumor effects, a 15% BSA/HA solution constituting the bottoms of BMNs was chosen to load TMZ, showing sustained drug release for over 28 days, guaranteeing long-term DNA damage in TMZ-resistant cells (U251-TR). Needle tips made from 10% BSA/HA solution loaded with NIC released the drug within 14 days, enhancing TMZ\'s efficacy by inhibiting the activity of O6-methylguanine-DNA methyltransferase (MGMT). BMNs exhibit superior mechanical properties, bypass the BBB, and gradually release the drug into the tumor periphery, thus significantly inhibiting tumor proliferation and expanding median survival in mice. The on-demand delivery of BMNs patches shows a strong translational potential for clinical applications, particularly in synergistic 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
    非小细胞肺癌(NSCLC)是发病率较高的主要疾病,生存率低,易对化疗产生耐药性。NSCLC化疗继发耐药的机制十分复杂,研究表明,STAT3(信号转导和转录激活因子3)的异常激活在其中起着重要作用。在这项研究中,以STAT3为精确靶标,构建了pGPU6/GFP/NeoSTAT3-shRNA重组质粒。通过在壳聚糖上修饰亲水和疏水嵌段,合成了多功能维生素E琥珀酸酯-壳聚糖-聚乙二醇单甲醚组氨酸(VES-CTS-mPEG-His)胶束。胶束可以通过自组装包封疏水性药物阿霉素,并通过正电荷和负电荷加载重组pGPU6/GFP/NeoSTAT3-shRNA(pDNA)以形成双负载纳米颗粒DOX/VCPH/pDNA。DOX和pDNA的共同递送和协同作用可以上调PTEN(磷酸酶和Tensin同源物)的表达,下调CD31的表达,诱导肿瘤细胞凋亡。精准靶向治疗结果显示,DOX/VCPH/pDNA可显著下调STAT3蛋白的表达水平,进一步增强化疗的疗效。通过这项研究,可以有效地实现NSCLC的精准个性化治疗,逆转其对化疗药物的耐药性,并为耐药NSCLC的治疗提供新的策略。
    Non-small cell lung cancer (NSCLC) is a major disease with high incidence, low survival rate and prone to develop drug resistance to chemotherapy. The mechanism of secondary drug resistance in NSCLC chemotherapy is very complex, and studies have shown that the abnormal activation of STAT3 (Signal Transducer and Activator of Transcription 3) plays an important role in it. In this study, the pGPU6/GFP/Neo STAT3-shRNA recombinant plasmid was constructed with STAT3 as the precise target. By modifying hydrophilic and hydrophobic blocks onto chitosan, a multifunctional vitamin E succinate-chitosan-polyethylene glycol monomethyl ether histidine (VES-CTS-mPEG-His) micelles were synthesized. The micelles could encapsulate hydrophobic drug doxorubicin through self-assembly, and load the recombinant pGPU6/GFP/Neo STAT3-shRNA (pDNA) through positive and negative charges to form dual-loaded nanoparticles DOX/VCPH/pDNA. The co-delivery and synergistic effect of DOX and pDNA could up-regulate the expression of PTEN (Phosphatase and Tensin Homolog), down-regulate the expression of CD31, and induce apoptosis of tumor cells. The results of precision targeted therapy showed that DOX/VCPH/pDNA could significantly down-regulate the expression level of STAT3 protein, further enhancing the efficacy of chemotherapy. Through this study, precision personalized treatment of NSCLC could be effectively achieved, reversing its resistance to chemotherapy drugs, and providing new strategies for the treatment of drug-resistant NSCLC.
    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
    缺氧和高浓度谷胱甘肽(GSH)严重妨碍了活性氧(ROS)在肿瘤医治中的感化和氧依附性战略。在这项工作中,建立了自生氧和自耗GSH透明质酸(HA)包被的卟啉纳米平台(TAPPP@CaO2/Pt(IV)/HA),用于增强肿瘤的光动力/离子/化学靶向协同治疗。在通过纳米系统进行ROS生产的过程中,实施GSH消耗策略以增强ROS诱导的氧化损伤,用于协同癌症治疗。纳米系统中的CaO2在酸性环境中分解成O2和H2O2,从而缓解缺氧并增强光动力治疗(PDT)效果。钙超载导致线粒体功能障碍并诱导细胞凋亡。Pt(IV)与GSH反应生成用于化疗的Pt(II)并降低GSH的浓度,保护ROS免于清除,以增强ROS诱导的氧化损伤。体外和体内结果表明,自我产生的氧气和自我消耗的GSH策略可以增强ROS依赖性PDT以及离子/化学协同疗法。所提出的策略不仅解决了缺氧限制PDT治疗效果的长期问题,而且还改善了肿瘤高度减少的环境。因此,TAPPP@CaO2/Pt(IV)/HA的制备为癌症的有效联合治疗提供了新的策略。
    Hypoxia and high concentration of glutathione (GSH) in tumor seriously hinder the role of reactive oxygen species (ROS) and oxygen-dependence strategy in tumor treatment. In this work, a self-generating oxygen and self-consuming GSH hyaluronic acid (HA)-coated porphyrin nanoplatform (TAPPP@CaO2/Pt(IV)/HA) is established for enhancing photodynamic/ion/chemo targeting synergistic therapy of tumor. During the efforts of ROS production by nanosystems, a GSH consuming strategy is implemented for augmenting ROS-induced oxidative damage for synergetic cancer therapy. CaO2 in the nanosystems is decomposed into O2 and H2O2 in an acidic environment, which alleviates hypoxia and enhances the photodynamic therapy (PDT) effect. Calcium overload causes mitochondria dysfunction and induces apoptosis. Pt (IV) reacts with GSH to produce Pt (II) for chemotherapy and reduce the concentration of GSH, protecting ROS from scavenging for augmenting ROS-induced oxidative damage. In vitro and in vivo results demonstrated the self-generating oxygen and self-consuming GSH strategy can enhance ROS-dependent PDT coupled with ion/chemo synergistic therapy. The proposed strategy not only solves the long-term problem that hypoxia limits therapeutic effect of PDT, but also ameliorates the highly reducing environment of tumors. Thus the preparation of TAPPP@CaO2/Pt(IV)/HA provided a novel strategy for the effective combined therapy of 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
    光热,光动力和声动力癌症治疗提供了精确的肿瘤消融和减少副作用的机会。干扰素基因的环状鸟苷酸腺苷酸合成酶刺激物(cGAS-STING)途径被认为是刺激患者免疫系统并实现持续免疫应答的潜在靶标。结合光热,使用cGAS-STING激动剂的光动力和声动力疗法代表了一种新开发的癌症治疗方法,在其对免疫系统的影响方面表现出明显的创新。最近的综述集中在各种材料及其在癌症治疗中的功能上。在这次审查中,我们专注于光热的分子机制,光动力和声动力癌症治疗以及cGAS-STING激动剂在治疗癌症中的相关作用。
    Photothermal, photodynamic and sonodynamic cancer therapies offer opportunities for precise tumor ablation and reduce side effects. The cyclic guanylate adenylate synthase-stimulator of interferon genes (cGAS-STING) pathway has been considered a potential target to stimulate the immune system in patients and achieve a sustained immune response. Combining photothermal, photodynamic and sonodynamic therapies with cGAS-STING agonists represents a newly developed cancer treatment demonstrating noticeable innovation in its impact on the immune system. Recent reviews have concentrated on diverse materials and their function in cancer therapy. In this review, we focus on the molecular mechanism of photothermal, photodynamic and sonodynamic cancer therapies and the connected role of cGAS-STING agonists in treating cancer.
    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
    由于人口老龄化,阿尔茨海默病(AD)的患病率在全球范围内呈上升趋势。然而,AD的有效临床治疗策略仍然难以捉摸。迄今为止,AD发病的潜在机制及其病理因素之间的相互作用尚不清楚。证据表明,AD进展最终是由神经元丢失驱动的,这又是由神经凋亡和神经炎症引起的。因此,抑制神经细胞凋亡和神经炎症可能是一种有用的抗AD策略。尽管如此,血脑屏障(BBB)阻碍了活性药物向脑实质的递送。为了应对这一挑战,我们制造了一种基于黑磷纳米片(BP)的亚甲蓝(MB)递送系统(BP-MB)用于AD治疗。确认成功制备BP-MB后,我们证明了在近红外光照射下其BBB交叉能力增强。体外药效学分析表明,BP和MB可以协同清除冈田酸(OA)处理的PC12细胞和脂多糖(LPS)处理的BV2细胞中过量的活性氧(ROS),从而有效逆转神经细胞凋亡和神经炎症。为了研究体内药效学,我们建立了AD小鼠模型,行为测试证实BP-MB治疗可以成功改善这些动物的认知功能。值得注意的是,病理评估结果与体外试验结果一致。结果表明,BP-MB可以清除过量的ROS并抑制Tau过度磷酸化,从而减轻下游神经细胞凋亡并调节小胶质细胞从促炎M1表型到抗炎M2表型的极化。总的来说,这项研究强调了具有逆转神经凋亡和神经炎症能力的智能纳米药物在AD治疗中的治疗潜力.
    The prevalence of Alzheimer\'s disease (AD) is increasing globally due to population aging. However, effective clinical treatment strategies for AD still remain elusive. The mechanisms underlying AD onset and the interplay between its pathological factors have so far been unclear. Evidence indicates that AD progression is ultimately driven by neuronal loss, which in turn is caused by neuroapoptosis and neuroinflammation. Therefore, the inhibition of neuroapoptosis and neuroinflammation could be a useful anti-AD strategy. Nonetheless, the delivery of active drug agents into the brain parenchyma is hindered by the blood-brain barrier (BBB). To address this challenge, we fabricated a black phosphorus nanosheet (BP)-based methylene blue (MB) delivery system (BP-MB) for AD therapy. After confirming the successful preparation of BP-MB, we proved that its BBB-crossing ability was enhanced under near-infrared light irradiation. In vitro pharmacodynamics analysis revealed that BP and MB could synergistically scavenge excessive reactive oxygen species (ROS) in okadaic acid (OA)-treated PC12 cells and lipopolysaccharide (LPS)-treated BV2 cells, thus efficiently reversing neuroapoptosis and neuroinflammation. To study in vivo pharmacodynamics, we established a mouse model of AD mice, and behavioral tests confirmed that BP-MB treatment could successfully improve cognitive function in these animals. Notably, the results of pathological evaluation were consistent with those of the in vitro assays. The findings demonstrated that BP-MB could scavenge excessive ROS and inhibit Tau hyperphosphorylation, thereby alleviating downstream neuroapoptosis and regulating the polarization of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Overall, this study highlights the therapeutic potential of a smart nanomedicine with the capability of reversing neuroapoptosis and neuroinflammation for AD 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
    目前,已经报道的少数关于视网膜母细胞瘤的光热/化疗研究主要限于涉及皮下植入的异位模型.然而,眼球是独特的生理结构,血-视网膜屏障(BRB)阻碍药物分子通过全身途径的吸收。此外,眼底丰富的血液循环加速药物代谢。为了维持所需的药物浓度,患者必须进行频繁的化疗.
    为了应对上述挑战,我们需要开发一种安全有效的眼底给药系统(FA-PEG-PDA-DOX)。
    我们提供了卓越的治疗功效,副作用最小或没有副作用,并成功建立了原位小鼠模型。我们评估了FA-PEG-PDA-DOX纳米系统的细胞摄取性能和靶向效率,并评估了其在体外和体内的协同抗肿瘤作用。进行生物分布评估以确定NPs在体内的保留时间和靶向效率。此外,进行了安全性评估。
    FA-PEG-PDA-DOX激光组的细胞内吞率为DOX组的5.23倍,是FA-PEG-PDA-DOX组的2.28倍。FA-PEG-PDA-DOX的荧光信号在肿瘤部位持续超过120小时。光热化疗组增殖周期内肿瘤细胞数量(17.2%)减少61.6%,与生理盐水对照组(78.8%)相反。FA-PEG-PDA-DOX纳米颗粒(NPs)具有良好的生物安全性和高生物相容性。
    双功能靶向纳米系统,在DOX和辐射温和升温的影响下,在裸鼠模型中产生精确的化学/光热疗法。
    UNASSIGNED: At present, the few photothermal/chemotherapy studies about retinoblastoma that have been reported are mainly restricted to ectopic models involving subcutaneous implantation. However, eyeball is unique physiological structure, the blood-retina barrier (BRB) hinders the absorption of drug molecules through the systemic route. Moreover, the abundant blood circulation in the fundus accelerates drug metabolism. To uphold the required drug concentration, patients must undergo frequent chemotherapy sessions.
    UNASSIGNED: To address these challenges above, we need to develop a secure and effective drug delivery system (FA-PEG-PDA-DOX) for the fundus.
    UNASSIGNED: We offered superior therapeutic efficacy with minimal or no side effects and successfully established orthotopic mouse models. We evaluated cellular uptake performance and targeting efficiency of FA-PEG-PDA-DOX nanosystem and assessed its synergistic antitumor effects in vitro and vivo. Biodistribution assessments were performed to determine the retention time and targeting efficiency of the NPs in vivo. Additionally, safety assessments were conducted.
    UNASSIGNED: Cell endocytosis rates of the FA-PEG-PDA-DOX+Laser group became 5.23 times that of the DOX group and 2.28 times that of FA-PEG-PDA-DOX group without irradiation. The fluorescence signal of FA-PEG-PDA-DOX persisted for more than 120 hours at the tumor site. The number of tumor cells (17.2%) in the proliferative cycle decreased by 61.6% in the photothermal-chemotherapy group, in contrast to that of the saline control group (78.8%). FA-PEG-PDA-DOX nanoparticles(NPs) exhibited favorable biosafety and high biocompatibility.
    UNASSIGNED: The dual functional targeted nanosystem, with the effects of DOX and mild-temperature elevation by irradiation, resulted in precise chemo/photothermal therapy in nude mice model.
    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
    增强肿瘤细胞内的氧化应激可以有效抑制三阴性乳腺癌(TNBC)的生长和转移。因此,可以有效破坏氧化还原平衡的创新纳米药物的开发代表了一种有希望但具有挑战性的TNBC治疗策略。在这项研究中,氧化应激放大器,表示为PBCH,包含PdAg介孔纳米酶和CaP矿化层,负载有GSH抑制剂L-丁硫氨酸亚砜胺(BSO),并引入可靶向CD44的透明质酸进一步表面修饰。在酸性肿瘤微环境中,Ca2+最初被释放,从而导致线粒体功能障碍并最终引发细胞凋亡。此外,BSO抑制细胞内还原的GSH的合成,并进一步放大癌细胞中的氧化应激水平。此外,PdAg纳米酶可以被近红外光激活以诱导光热和光动力效应,引起ROS爆发,同时通过引起免疫原性细胞死亡促进细胞凋亡。PBCH的高性能治疗效果,基于上述多重氧化损伤和光热消融的协同作用,在TNBC细胞和动物模型中得到验证,宣布其作为一种安全有效的抗肿瘤药物的潜力。所提出的方法为精确有效地治疗TNBC提供了新的视角。
    Amplifying oxidative stress within tumor cells can effectively inhibit the growth and metastasis of triple-negative breast cancer (TNBC). Therefore, the development of innovative nanomedicines that can effectively disrupt the redox balance represents a promising yet challenging therapeutic strategy for TNBC. In this study, an oxidative stress amplifier, denoted as PBCH, comprising PdAg mesoporous nanozyme and a CaP mineralized layer, loaded with GSH inhibitor L-buthionine sulfoximine (BSO), and further surface-modified with hyaluronic acid that can target CD44, is introduced. In the acidic tumor microenvironment, Ca2+ is initially released, thereby leading to mitochondrial dysfunction and eventually triggering apoptosis. Additionally, BSO suppresses the synthesis of intracellular reduced GSH and further amplifies the level of oxidative stress in cancer cells. Furthermore, PdAg nanozyme can be activated by near-infrared light to induce photothermal and photodynamic effects, causing a burst of ROS and simultaneously promoting cell apoptosis via provoking immunogenic cell death. The high-performance therapeutic effects of PBCH, based on the synergistic effect of aforementioned multiple oxidative damage and photothermal ablation, are validated in TNBC cells and animal models, declaring its potential as a safe and effective anti-tumor agent. The proposed approach offers new perspectives for precise and efficient treatment of TNBC.
    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
    由于肿瘤微环境(TME)中谷胱甘肽(GSH)过多和缺氧,单独的化学动力学疗法(CDT)无法达到足够的治疗效果。迫切需要制定一种新的策略来提高效率。在这里,我们制备了由胶体二氧化硅衍生的硅酸铜纳米平台(CSNP)。CSNP中的Cu(II)可以还原成Cu(I),级联以诱导随后的CDT过程。此外,受益于660nm激光照射下的GSH消耗和氧气(O2)产生,CSNP表现出Fenton样和缺氧缓解活性,有助于在TME中有效产生超氧阴离子自由基(•O2-)和羟基自由基(•OH)。此外,具有合适的带隙特性和优异的光化学性质,CSNP还可以用作光动力疗法(PDT)的有效I型光敏剂。CSNP的协同CDT/PDT活性在体外和体内实验中均表现出有效的抗肿瘤作用和生物安全性。集成Fenton样和光敏特性的一体化纳米平台的开发可以改善肿瘤内的ROS产生。这项研究强调了硅酸盐纳米材料在癌症治疗中的潜力。
    Chemodynamic therapy (CDT) alone cannot achieve sufficient therapeutic effects due to the excessive glutathione (GSH) and hypoxia in the tumor microenvironment (TME). Developing a novel strategy to improve efficiency is urgently needed. Herein, we prepared a copper silicate nanoplatform (CSNP) derived from colloidal silica. The Cu(II) in CSNP can be reduced to Cu(I), which cascades to induce a subsequent CDT process. Additionally, benefiting from GSH depletion and oxygen (O2) generation under 660 nm laser irradiation, CSNP exhibits both Fenton-like and hypoxia-alleviating activities, contributing to the effective generation of superoxide anion radical (•O2-) and hydroxyl radical (•OH) in the TME. Furthermore, given the suitable band-gap characteristic and excellent photochemical properties, CSNP can also serve as an efficient type-I photosensitizer for photodynamic therapy (PDT). The synergistic CDT/PDT activity of CSNP presents an efficient antitumor effect and biosecurity in both in vitro and in vivo experiments. The development of an all-in-one nanoplatform that integrates Fenton-like and photosensing properties could improve ROS production within tumors. This study highlights the potential of silicate nanomaterials 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
    克罗恩病(CD)是一种病因不明的难治性慢性炎症性肠病(IBD)。透壁性炎症,涉及肠道和肠系膜,代表CD的特征性病理特征,并且是其难处理性的关键因素。这里,这项研究描述了一种载有肿瘤坏死因子-α(TNF-α)脱氧核酶(DNAzymes)(DNAzymes@可降解硅纳米颗粒@甘露糖,Dz@MDSN),它可以在炎症部位靶向巨噬细胞,并对活性氧(ROS)产生反应以释放药物。Dz@MDSN不仅可以通过降解TNF-αmRNA来打破巨噬细胞的炎症循环,而且可以减少主要来自巨噬细胞的ROS的产生。此外,Dz@MDSN通过清除ROS抑制上皮细胞过度的焦亡,从而修复肠道屏障,减少肠道细菌向肠系膜的易位。因此,这些联合作用协同作用有助于抑制肠道和肠系膜内的炎症。这项研究可能代表了利用纳米材料实现CD的透壁愈合领域的首次成功尝试。这也为CD患者提供了一种有希望的治疗策略。
    Crohn\'s disease (CD) is a refractory chronic inflammatory bowel disease (IBD) with unknown etiology. Transmural inflammation, involving the intestine and mesentery, represents a characteristic pathological feature of CD and serves as a critical contributor to its intractability. Here, this study describes an oral pyroptosis nanoinhibitor loaded with tumor necrosis factor-α (TNF-α) deoxyribozymes (DNAzymes) (DNAzymes@degradable silicon nanoparticles@Mannose, Dz@MDSN), which can target macrophages at the site of inflammation and respond to reactive oxygen species (ROS) to release drugs. Dz@MDSN can not only break the inflammatory cycle in macrophages by degrading TNF-α mRNA but also reduce the production of ROS mainly from macrophages. Moreover, Dz@MDSN inhibits excessive pyroptosis in epithelial cells through ROS clearance, thereby repairing the intestinal barrier and reducing the translocation of intestinal bacteria to the mesentery. Consequently, these combined actions synergistically contribute to the suppression of inflammation within both the intestine and the mesentery. This study likely represents the first successful attempt in the field of utilizing nanomaterials to achieve transmural healing for CD, which also provides a promising treatment strategy for CD patients.
    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
    铂(II)药物作为一线抗癌试剂受到副作用和耐药性的限制。刺激响应性纳米系统有望对药物输送进行精确的时空操纵,目的是提高生物利用度和减少副作用。在这里,具有十八烷基脂肪链和组蛋白脱乙酰酶抑制剂的多靶向八面体铂(IV)前药(苯基丁酸,PHB)在轴向位置以改善顺铂的治疗效果,通过疏水相互作用负载在上转换纳米颗粒(UCNP)上。随后的DSPE-PEG2000和精氨酸-甘氨酸-天冬氨酸(RGD)肽的连接赋予了纳米载体高的生物相容性和肿瘤特异性。制造的纳米粒子(UCNP/Pt(IV)-RGD)可以通过上转换发光(UCL)辐照和谷胱甘肽(GSH)还原来触发,以可控地释放Pt(II)物种和PHB,诱导深刻的细胞毒性。体外和体内实验均表明UCNP/Pt(IV)-RGD具有显著的抗肿瘤效果,高肿瘤靶向特异性,和实时UCL成像能力,提出了一种智能的铂(IV)前药纳米载体,用于UCL引导的双重刺激反应性联合治疗。
    Platinum(II) drugs as a first-line anticancer reagent are limited by side effects and drug resistance. Stimuli-responsive nanosystems hold promise for precise spatiotemporal manipulation of drug delivery, with the aim to promote bioavailability and minimize side effects. Herein, a multitargeting octahedral platinum(IV) prodrug with octadecyl aliphatic chain and histone deacetylase inhibitor (phenylbutyric acid, PHB) at axial positions to improve the therapeutic effect of cisplatin was loaded on the upconversion nanoparticles (UCNPs) through hydrophobic interaction. Followed attachment of DSPE-PEG2000 and arginine-glycine-aspartic (RGD) peptide endowed the nanovehicles with high biocompatibility and tumor specificity. The fabricated nanoparticles (UCNP/Pt(IV)-RGD) can be triggered by upconversion luminescence (UCL) irradiation and glutathione (GSH) reduction to controllably release Pt(II) species and PHB, inducing profound cytotoxicity. Both in vitro and in vivo experiments demonstrated that UCNP/Pt(IV)-RGD exhibited remarkable antitumor efficiency, high tumor-targeting specificity, and real-time UCL imaging capacity, presenting an intelligent platinum(IV) prodrug-loaded nanovehicle for UCL-guided dual-stimuli-responsive combination therapy.
    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