首页 > 最新文献

Nano Today最新文献

英文 中文
Construction of N-Fe-S bridge in atomic iron catalyst for boosting Fenton-like reactions 在原子铁催化剂中构建 N-Fe-S 桥,促进类似芬顿的反应
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-22 DOI: 10.1016/j.nantod.2024.102462

Constructing the coordination environment of single-atom catalysts (SACs) can be an attractive technical strategy to regulate the generation of reactive oxygen species for Fenton-like reactions. Herein, we have constructed an asymmetric coordination of Fe single-atom catalyst (FeSA-NS-PCNS) with abundant N-Fe-S bridge (FeSA-N3S1) for robust Fenton-like reactions. 82.5 % of singlet oxygen (1O2) selectivity and high turnover frequency of bisphenol A degradation (0.568 min−1) were achieved at mild conditions. Experimental works and theoretical analyses illustrated that S doping breaks the inert environment of the original N-Fe-N symmetric coordination equilibrium and modulates the electron density of the atomic Fe center, which is beneficial for boosting PMS adsorption and reducing the energy barriers of vital *OH and *O intermediates. The coupling between the FeSA-N3S1 interface and peroxymonosulfate molecule boosts in-situ electron transfer through the N-Fe-S bridge, which induces more electron flow from the low valence Fe to OH* on the surface of Fe-*O-H, forming a high yield of 1O2. Moreover, we designed the Fenton-like reactions by FeSA-NS-PCNS membrane reactor for an efficient contaminant removal rate of over 90 % even after 11 cycles. This work provides a novel perspective on developing SACs with asymmetric coordination to regulate reactive oxygen species for the treatment of organic contaminants in water bodies.

构建单原子催化剂(SACs)的配位环境是调节芬顿类反应中活性氧生成的一种极具吸引力的技术策略。在此,我们构建了一种具有丰富 N-Fe-S 桥(FeSA-N3S1)的不对称配位铁单原子催化剂(FeSA-NS-PCNS),用于强健的类芬顿反应。在温和的条件下,单线态氧(1O2)的选择性达到了 82.5%,双酚 A 的降解周转频率也很高(0.568 min-1)。实验工作和理论分析表明,S 掺杂打破了原有 N-Fe-N 对称配位平衡的惰性环境,调节了原子 Fe 中心的电子密度,有利于促进 PMS 的吸附,降低重要 *OH 和 *O 中间体的能垒。FeSA-N3S1 界面与过一硫酸分子之间的耦合促进了通过 N-Fe-S 桥的原位电子转移,从而诱导更多的电子从低价位的 Fe 流向 Fe-*O-H 表面的 OH*,形成高产率的 1O2。此外,我们还通过 FeSA-NS-PCNS 膜反应器设计了类似芬顿的反应,即使经过 11 次循环,污染物去除率也能达到 90% 以上。这项工作为开发具有不对称配位的 SACs 提供了一个新的视角,以调节水体中有机污染物处理过程中的活性氧物种。
{"title":"Construction of N-Fe-S bridge in atomic iron catalyst for boosting Fenton-like reactions","authors":"","doi":"10.1016/j.nantod.2024.102462","DOIUrl":"10.1016/j.nantod.2024.102462","url":null,"abstract":"<div><p>Constructing the coordination environment of single-atom catalysts (SACs) can be an attractive technical strategy to regulate the generation of reactive oxygen species for Fenton-like reactions. Herein, we have constructed an asymmetric coordination of Fe single-atom catalyst (Fe<sub>SA</sub>-NS-PCNS) with abundant N-Fe-S bridge (Fe<sub>SA</sub>-N<sub>3</sub>S<sub>1</sub>) for robust Fenton-like reactions. 82.5 % of singlet oxygen (<sup>1</sup>O<sub>2</sub>) selectivity and high turnover frequency of bisphenol A degradation (0.568 min<sup>−1</sup>) were achieved at mild conditions. Experimental works and theoretical analyses illustrated that S doping breaks the inert environment of the original N-Fe-N symmetric coordination equilibrium and modulates the electron density of the atomic Fe center, which is beneficial for boosting PMS adsorption and reducing the energy barriers of vital *OH and *O intermediates. The coupling between the Fe<sub>SA</sub>-N<sub>3</sub>S<sub>1</sub> interface and peroxymonosulfate molecule boosts in-situ electron transfer through the N-Fe-S bridge, which induces more electron flow from the low valence Fe to OH* on the surface of Fe-*O-H, forming a high yield of <sup>1</sup>O<sub>2</sub>. Moreover, we designed the Fenton-like reactions by Fe<sub>SA</sub>-NS-PCNS membrane reactor for an efficient contaminant removal rate of over 90 % even after 11 cycles. This work provides a novel perspective on developing SACs with asymmetric coordination to regulate reactive oxygen species for the treatment of organic contaminants in water bodies.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142040365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Iron consumption strengthens anti-tumoral STING activation mediated by manganese-based nanoparticles 消耗铁能增强锰基纳米粒子介导的 STING 抗肿瘤激活作用
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-22 DOI: 10.1016/j.nantod.2024.102446

The cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway, as an important part in innate immunity, has recently emerged as a promising target for improving tumor therapy. Manganese ions (Mn2+) are an emerging agonist in the cGAS-STING pathway with multifaceted advantages, however manganese-based nanoparticles alone as the Mn2+ source have shown limited activity in eliciting anti-tumor immune responses compared to conventional organic STING agonists, and the underlying mechanism of the suboptimal efficiency remains unclear. Here, we demonstrate that intratumoral iron ions attenuate manganese-induced anti-tumor STING activation, and that the utilization of deferoxamine (DFO), an iron chelator that depletes intratumoral iron ions, effectively increases the intracellular accumulation of Mn2+ and thus promoted the STING activation efficiency of a hyaluronic acid modified manganese carbonate-silica hybrid nanoparticle (DS@Mn-H) in macrophages. The mechanism study suggests that the addition of DFO inhibited the expression of ferroportin (FPN), which serves as a Mn2+ exporter to reduce intracellular Mn2+ level. The synergistic effect of DS@Mn-H and DFO achieved excellent anti-tumor activities in a mouse colon carcinoma model. This work provides new insights on improving the Mn-based metallo-immunotherapy of cancer.

环GMP-AMP合成酶(cGAS)/干扰素基因刺激器(STING)通路是先天性免疫的重要组成部分,最近已成为改善肿瘤治疗的一个很有前景的靶点。锰离子(Mn2+)是cGAS-STING通路中一种新兴的激动剂,具有多方面的优势,但与传统的有机STING激动剂相比,仅以锰基纳米粒子作为Mn2+源在激发抗肿瘤免疫反应方面的活性有限,而且效率不佳的内在机制仍不清楚。在这里,我们证明了瘤内铁离子会减弱锰诱导的抗肿瘤 STING 激活,而利用去铁胺(DFO)这种能消耗瘤内铁离子的铁螯合剂,能有效增加 Mn2+ 在细胞内的积累,从而促进透明质酸修饰的碳酸锰-二氧化硅杂化纳米粒子(DS@Mn-H)在巨噬细胞中的 STING 激活效率。机理研究表明,DFO的加入抑制了铁蛋白(FPN)的表达,而铁蛋白是Mn2+的输出者,可降低细胞内的Mn2+水平。在小鼠结肠癌模型中,DS@Mn-H 和 DFO 的协同作用取得了优异的抗肿瘤活性。这项工作为改进基于锰的癌症金属免疫疗法提供了新的见解。
{"title":"Iron consumption strengthens anti-tumoral STING activation mediated by manganese-based nanoparticles","authors":"","doi":"10.1016/j.nantod.2024.102446","DOIUrl":"10.1016/j.nantod.2024.102446","url":null,"abstract":"<div><p>The cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway, as an important part in innate immunity, has recently emerged as a promising target for improving tumor therapy. Manganese ions (Mn<sup>2+</sup>) are an emerging agonist in the cGAS-STING pathway with multifaceted advantages, however manganese-based nanoparticles alone as the Mn<sup>2+</sup> source have shown limited activity in eliciting anti-tumor immune responses compared to conventional organic STING agonists, and the underlying mechanism of the suboptimal efficiency remains unclear. Here, we demonstrate that intratumoral iron ions attenuate manganese-induced anti-tumor STING activation, and that the utilization of deferoxamine (DFO), an iron chelator that depletes intratumoral iron ions, effectively increases the intracellular accumulation of Mn<sup>2+</sup> and thus promoted the STING activation efficiency of a hyaluronic acid modified manganese carbonate-silica hybrid nanoparticle (DS@Mn-H) in macrophages. The mechanism study suggests that the addition of DFO inhibited the expression of ferroportin (FPN), which serves as a Mn<sup>2+</sup> exporter to reduce intracellular Mn<sup>2+</sup> level. The synergistic effect of DS@Mn-H and DFO achieved excellent anti-tumor activities in a mouse colon carcinoma model. This work provides new insights on improving the Mn-based metallo-immunotherapy of cancer.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1748013224003025/pdfft?md5=9b8d2031c4e24675decc08d04016ae5a&pid=1-s2.0-S1748013224003025-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142045034","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Injectable thermosensitive hydrogels loaded with irradiated tumor cell-derived microparticles and manganese activate anti-tumor immunity 可注射的热敏性水凝胶装载了辐照肿瘤细胞衍生的微颗粒和锰,可激活抗肿瘤免疫力
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-22 DOI: 10.1016/j.nantod.2024.102455

Immunosuppressive tumor microenvironment and inadequate activation of the innate immune system are important reasons for the failure of systemic anti-tumor immunotherapy. Irradiated tumor cell-derived microparticles (RMPs) can induce immunogenic cell death (ICD) of tumor cells and inhibit the M2-like phenotype of tumor-associated macrophages (TAMs). The current study found that double-stranded DNA was enriched in RMPs, which combined with manganese ions (Mn2+), the natural activator of cGAS-STING signaling, and synergistically amplified cGAS-STING signaling cascade in antigen-presenting cells (APCs). On this basis, a polyamino acid thermosensitive hydrogel delivery system was designed to simultaneously load RMPs and Mn2+. The obtained RMPs@Mn2+ hydrogel exhibited thermosensitivity, biocompatibility and sustained release characteristics. The intratumoral injection of RMPs@Mn2+ slowly released RMPs and Mn2+, inducing ICD of tumor cells, continuously activating cGAS-STING signaling of APCs, reprograming TAMs to M1-like phenotype, and promoting the activation of dendritic cells in tumor draining lymph nodes. The full activation of innate immunity at the tumor site further promoted priming and tumor infiltration of T cells, leading to tumor regression. RMPs@Mn2+ combined with anti-PD-1 achieved strong anti-cancer efficacy in a refractory malignant ascites model, in which more than 50 % of mice with malignant ascites achieved complete regression and long-term immune memory.

免疫抑制性肿瘤微环境和先天性免疫系统激活不足是全身性抗肿瘤免疫疗法失败的重要原因。经过辐照的肿瘤细胞衍生微颗粒(RMPs)可以诱导肿瘤细胞的免疫原性细胞死亡(ICD),并抑制肿瘤相关巨噬细胞(TAMs)的M2样表型。目前的研究发现,RMPs 中富含双链 DNA,它与 cGAS-STING 信号的天然激活剂锰离子(Mn2+)结合,协同放大了抗原递呈细胞(APCs)中的 cGAS-STING 信号级联。在此基础上,我们设计了一种聚氨基酸热敏水凝胶递送系统,可同时负载 RMPs 和 Mn2+。获得的 RMPs@Mn2+ 水凝胶具有热敏性、生物相容性和持续释放特性。瘤内注射RMPs@Mn2+可缓慢释放RMPs和Mn2+,诱导肿瘤细胞ICD,持续激活APCs的cGAS-STING信号,将TAMs重编程为M1样表型,并促进肿瘤引流淋巴结中树突状细胞的活化。肿瘤部位先天性免疫的全面激活进一步促进了 T 细胞的引诱和肿瘤浸润,从而导致肿瘤消退。在难治性恶性腹水模型中,RMPs@Mn2+与抗-PD-1联用取得了很强的抗癌疗效,50%以上的恶性腹水小鼠实现了完全消退和长期免疫记忆。
{"title":"Injectable thermosensitive hydrogels loaded with irradiated tumor cell-derived microparticles and manganese activate anti-tumor immunity","authors":"","doi":"10.1016/j.nantod.2024.102455","DOIUrl":"10.1016/j.nantod.2024.102455","url":null,"abstract":"<div><p>Immunosuppressive tumor microenvironment and inadequate activation of the innate immune system are important reasons for the failure of systemic anti-tumor immunotherapy. Irradiated tumor cell-derived microparticles (RMPs) can induce immunogenic cell death (ICD) of tumor cells and inhibit the M2-like phenotype of tumor-associated macrophages (TAMs). The current study found that double-stranded DNA was enriched in RMPs, which combined with manganese ions (Mn<sup>2+</sup>), the natural activator of cGAS-STING signaling, and synergistically amplified cGAS-STING signaling cascade in antigen-presenting cells (APCs). On this basis, a polyamino acid thermosensitive hydrogel delivery system was designed to simultaneously load RMPs and Mn<sup>2+</sup>. The obtained RMPs@Mn<sup>2+</sup> hydrogel exhibited thermosensitivity, biocompatibility and sustained release characteristics. The intratumoral injection of RMPs@Mn<sup>2+</sup> slowly released RMPs and Mn<sup>2+</sup>, inducing ICD of tumor cells, continuously activating cGAS-STING signaling of APCs, reprograming TAMs to M1-like phenotype, and promoting the activation of dendritic cells in tumor draining lymph nodes. The full activation of innate immunity at the tumor site further promoted priming and tumor infiltration of T cells, leading to tumor regression. RMPs@Mn<sup>2+</sup> combined with anti-PD-1 achieved strong anti-cancer efficacy in a refractory malignant ascites model, in which more than 50 % of mice with malignant ascites achieved complete regression and long-term immune memory.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142045035","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Structural evolution and self-reconstruction of nickel hexacyanoferrate Prussian blue analogues for long-lasting ampere-current seawater oxidation 六氰基铁酸镍普鲁士蓝类似物的结构演化和自我重构,用于长效安培电流海水氧化
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-21 DOI: 10.1016/j.nantod.2024.102454

The commercial success of hydrogen (H2) production via seawater electrolysis largely depends on overcoming anode corrosion caused by chloride (Cl), a critical challenge for scaling up renewable energy storage. Herein, we present the synthesis of nickel hexacyanoferrate Prussian blue analogues via anodic polarization for efficient alkaline seawater oxidation (ASO). Electrochemical experiments and in situ Raman studies unveil that leaching of Fe(CN)63− from the electrode facilitates a dynamic self-reconstruction process and leads to the formation of high-valence metal reaction specie, and appropriate Fe(CN)63− effectively shields active sites from Cl interference even at ampere-level current density (j). In experiments, our electrode demonstrates an overpotential of merely 349 mV to reach a j of 1 A cm−2 and maintains stable operation for over 1000 h with protective Fe(CN)63−. This work showcases the potential of Prussian blue analogues for efficient and long-lasting ASO, protecting against Cl in large-scale seawater H2 electrolysis.

通过海水电解生产氢气(H2)的商业成功在很大程度上取决于能否克服氯化物(Cl-)引起的阳极腐蚀,这是扩大可再生能源储存规模所面临的关键挑战。在此,我们介绍了通过阳极极化合成六氰基铁酸镍普鲁士蓝类似物,用于高效碱性海水氧化(ASO)。电化学实验和原位拉曼研究揭示了电极中 Fe(CN)63- 的浸出促进了一个动态的自我重构过程,并导致高价金属反应物的形成,即使在安培级电流密度 (j) 下,适当的 Fe(CN)63- 也能有效屏蔽活性位点受到 Cl- 的干扰。在实验中,我们的电极仅需 349 mV 的过电位就能达到 1 A cm-2 的电流密度,并能在有 Fe(CN)63- 保护的情况下稳定工作 1000 小时以上。这项工作展示了普鲁士蓝类似物在大规模海水 H2 电解中用于高效、长效 ASO 以及抵御 Cl- 的潜力。
{"title":"Structural evolution and self-reconstruction of nickel hexacyanoferrate Prussian blue analogues for long-lasting ampere-current seawater oxidation","authors":"","doi":"10.1016/j.nantod.2024.102454","DOIUrl":"10.1016/j.nantod.2024.102454","url":null,"abstract":"<div><p>The commercial success of hydrogen (H<sub>2</sub>) production via seawater electrolysis largely depends on overcoming anode corrosion caused by chloride (Cl<sup>−</sup>), a critical challenge for scaling up renewable energy storage. Herein, we present the synthesis of nickel hexacyanoferrate Prussian blue analogues via anodic polarization for efficient alkaline seawater oxidation (ASO). Electrochemical experiments and in situ Raman studies unveil that leaching of Fe(CN)<sub>6</sub><sup>3−</sup> from the electrode facilitates a dynamic self-reconstruction process and leads to the formation of high-valence metal reaction specie, and appropriate Fe(CN)<sub>6</sub><sup>3−</sup> effectively shields active sites from Cl<sup>−</sup> interference even at ampere-level current density (<em>j</em>). In experiments, our electrode demonstrates an overpotential of merely 349 mV to reach a <em>j</em> of 1 A cm<sup>−2</sup> and maintains stable operation for over 1000 h with protective Fe(CN)<sub>6</sub><sup>3−</sup>. This work showcases the potential of Prussian blue analogues for efficient and long-lasting ASO, protecting against Cl<sup>−</sup> in large-scale seawater H<sub>2</sub> electrolysis.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142021044","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The emerging role of nanozymes in ocular antioxidant therapy 纳米酶在眼部抗氧化疗法中的新兴作用
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-21 DOI: 10.1016/j.nantod.2024.102448

Oxidative stress significantly contributes to various ocular diseases, necessitating innovative therapeutic approaches. This manuscript explores nanozymes which are nanomaterials with enzyme-like activities, as potential therapeutic agents for oxidative stress-related eye diseases. The rising global incidence of visual impairments underscores the urgent requirement for effective, adaptable, and safe therapeutic options. We detail how oxidative stress damages ocular tissues, the limitations of natural antioxidants, and discuss the benefits of nanozymes, such as their stability, multi-enzyme activity, and enhanced drug delivery capabilities. Our review covers nanozyme applications in various eye diseases, including diabetic retinopathy, age-related macular degeneration, retinopathy of prematurity, glaucoma, optic neuritis, cataracts, dry eye syndrome, chemical corneal burns, and uveitis. We also explore innovative delivery methods, such as microneedles, contact lenses, and hydrogels, which can enhance the bioavailability and therapeutic efficacy of nanozymes. The manuscript concludes by emphasizing the existing limitations and the promising future of nanozymes in ocular antioxidant therapy. With their multi-enzyme activity and improved delivery systems, nanozymes represent a significant advancement over traditional treatments, offering new hope for managing and potentially curing various oxidative stress-related eye diseases.

氧化应激是各种眼部疾病的重要诱因,因此需要创新的治疗方法。本手稿探讨了纳米酶,即具有类似酶活性的纳米材料,作为治疗氧化应激相关眼病的潜在药物。全球视力损伤发病率的不断上升凸显了对有效、适应性强且安全的治疗方案的迫切需求。我们详细介绍了氧化应激如何损害眼部组织、天然抗氧化剂的局限性,并讨论了纳米酶的优点,如稳定性、多酶活性和更强的给药能力。我们的综述涵盖了纳米酶在各种眼病中的应用,包括糖尿病视网膜病变、老年性黄斑变性、早产儿视网膜病变、青光眼、视神经炎、白内障、干眼症、化学性角膜烧伤和葡萄膜炎。我们还探讨了创新的给药方法,如微针、隐形眼镜和水凝胶,它们可以提高纳米酶的生物利用度和疗效。手稿最后强调了纳米酶在眼部抗氧化疗法中的现有局限性和广阔前景。纳米酶具有多酶活性并改进了给药系统,是传统疗法的一大进步,为控制和治疗各种氧化应激相关眼病带来了新希望。
{"title":"The emerging role of nanozymes in ocular antioxidant therapy","authors":"","doi":"10.1016/j.nantod.2024.102448","DOIUrl":"10.1016/j.nantod.2024.102448","url":null,"abstract":"<div><p>Oxidative stress significantly contributes to various ocular diseases, necessitating innovative therapeutic approaches. This manuscript explores nanozymes which are nanomaterials with enzyme-like activities, as potential therapeutic agents for oxidative stress-related eye diseases. The rising global incidence of visual impairments underscores the urgent requirement for effective, adaptable, and safe therapeutic options. We detail how oxidative stress damages ocular tissues, the limitations of natural antioxidants, and discuss the benefits of nanozymes, such as their stability, multi-enzyme activity, and enhanced drug delivery capabilities. Our review covers nanozyme applications in various eye diseases, including diabetic retinopathy, age-related macular degeneration, retinopathy of prematurity, glaucoma, optic neuritis, cataracts, dry eye syndrome, chemical corneal burns, and uveitis. We also explore innovative delivery methods, such as microneedles, contact lenses, and hydrogels, which can enhance the bioavailability and therapeutic efficacy of nanozymes. The manuscript concludes by emphasizing the existing limitations and the promising future of nanozymes in ocular antioxidant therapy. With their multi-enzyme activity and improved delivery systems, nanozymes represent a significant advancement over traditional treatments, offering new hope for managing and potentially curing various oxidative stress-related eye diseases.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142012224","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cascade piezocatalytic nanoprodrug for synergistic piezocatalytic therapy and sono-activated chemotherapy-augmented immunotherapy 级联压触催化纳米药物,用于协同压触催化疗法和声激活化疗增强免疫疗法
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-20 DOI: 10.1016/j.nantod.2024.102453

Immune checkpoint blockade (ICB) has shown great promise in the management of various cancers. However, only a small percentage of patients profit from ICB therapy. Immunogenic cell death (ICD) has demonstrated its specific capability in reconstructing the tumor microenvironment (TME) and activating anti-tumor immunity. Herein, an ICD cascade enhancement based on BPTL nanoreactors is proposed to overcome the inadequate damage-associated molecular patterns of ICD inducers. BPTL nanoreactor is formed by incorporating the piezocatalytic BaTiO3 (T-BTO) nanoparticles and paclitaxel (PTX) prodrug that responds to reactive oxygen species (ROS) into liposome nanoparticles for synergistic piezocatalytic and sono-activated chemotherapy (SACT)-augmented immunotherapy. Ultrasound (US) irradiation of the designed BPTL initiates a superior piezodynamic effect and produces ROS by piezocatalytic therapy to trigger ICD. Subsequently, the generated ROS breaks up the thioketal bonds in BPTL, thereby leading to the on-demand release of PTX for SACT and augmented ICD activation. In vivo evaluation demonstrated that BTPL with US irradiation significantly eradicated primary and distant metastatic tumors and markedly prevented the lung metastasis. This nanoplatform combines US-triggered piezocatalytic therapy and SACT for ICD stimulation, providing a robust strategy for amplifying piezocatalytic effect-mediated immune response against tumors.

免疫检查点阻断疗法(ICB)在治疗各种癌症方面显示出巨大的前景。然而,只有一小部分患者能从 ICB 疗法中获益。免疫原性细胞死亡(ICD)在重建肿瘤微环境(TME)和激活抗肿瘤免疫方面显示了其特殊能力。本文提出了一种基于BPTL纳米反应器的ICD级联增强技术,以克服ICD诱导剂损伤相关分子模式不足的问题。BPTL纳米反应器是将压催化BaTiO3(T-BTO)纳米颗粒和对活性氧(ROS)有反应的紫杉醇(PTX)原药加入脂质体纳米颗粒中形成的,用于协同压催化和声激活化疗(SACT)增强免疫疗法。用超声波(US)照射设计好的 BPTL 可产生卓越的压动力效应,并通过压催化疗法产生 ROS,从而触发 ICD。随后,产生的 ROS 会破坏 BPTL 中的硫酮键,从而按需释放用于 SACT 的 PTX,并增强 ICD 的激活。体内评估结果表明,BPTL 经 US 照射后可显著消除原发性和远处转移性肿瘤,并明显防止肺转移。该纳米平台将 US 触发的压电催化疗法和 SACT 刺激 ICD 结合在一起,为扩大压电催化效应介导的抗肿瘤免疫反应提供了一种强有力的策略。
{"title":"Cascade piezocatalytic nanoprodrug for synergistic piezocatalytic therapy and sono-activated chemotherapy-augmented immunotherapy","authors":"","doi":"10.1016/j.nantod.2024.102453","DOIUrl":"10.1016/j.nantod.2024.102453","url":null,"abstract":"<div><p>Immune checkpoint blockade (ICB) has shown great promise in the management of various cancers. However, only a small percentage of patients profit from ICB therapy. Immunogenic cell death (ICD) has demonstrated its specific capability in reconstructing the tumor microenvironment (TME) and activating anti-tumor immunity. Herein, an ICD cascade enhancement based on BPTL nanoreactors is proposed to overcome the inadequate damage-associated molecular patterns of ICD inducers. BPTL nanoreactor is formed by incorporating the piezocatalytic BaTiO<sub>3</sub> (T-BTO) nanoparticles and paclitaxel (PTX) prodrug that responds to reactive oxygen species (ROS) into liposome nanoparticles for synergistic piezocatalytic and sono-activated chemotherapy (SACT)-augmented immunotherapy. Ultrasound (US) irradiation of the designed BPTL initiates a superior piezodynamic effect and produces ROS by piezocatalytic therapy to trigger ICD. Subsequently, the generated ROS breaks up the thioketal bonds in BPTL, thereby leading to the on-demand release of PTX for SACT and augmented ICD activation. In vivo evaluation demonstrated that BTPL with US irradiation significantly eradicated primary and distant metastatic tumors and markedly prevented the lung metastasis. This nanoplatform combines US-triggered piezocatalytic therapy and SACT for ICD stimulation, providing a robust strategy for amplifying piezocatalytic effect-mediated immune response against tumors.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142012223","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dynamic polyphenol nanoparticles boost cuproptosis-driven metalloimmunotherapy of breast cancer 动态多酚纳米粒子促进乳腺癌的杯突症金属免疫疗法
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-17 DOI: 10.1016/j.nantod.2024.102442

Polyphenolic materials have been extensively explored for biomedical applications. However, most polymer-based polyphenols are synthesized by covalently grafting phenol groups on the backbone of polymers and more convenient synthetic strategy remains challenging. Herein, a general and robust strategy to synthesize polypeptide-based polyphenol via selective ortho-hydroxylation of poly(tyrosine) was developed to construct dynamic nanoparticles (GPCuD NPs) composed of Cu2+ and phenylboronic acid modified doxorubicin prodrug (DOX-PBA) via metal-phenol coordination interaction and pH-reversible phenylboronate ester bond, respectively. Furthermore, matrix metalloproteinase-2 (MMP-2)-cleavable GPLGLAG peptide was also included between the polyethylene glycol and polyphenol segments, endowing GPCuD NPs with tumor-specific accumulation and deep penetration through enzyme-triggered depegylation. Notably, Cu2+-chelated nanoparticles efficiently ameliorated the immunosuppressive tumor microenvironment via recruiting antitumor immune cells and repolarizing M2-type tumor-associated macrophages to M1 phenotype. The combination of cuproptosis-driven metalloimmunotherapy with DOX chemotherapy remarkably suppressed 4T1 orthotopic breast tumor growth by 81.9 % and established a long-term immune memory (effective memory T cells up to 30.8 %) to prevent lung metastasis. This study has demonstrated a generalizable polyphenol nanoplatform for tumor-targeted and cuproptosis-regulated combination cancer metalloimmunotherapy.

多酚材料在生物医学领域的应用已得到广泛探索。然而,大多数聚合物基多酚都是通过在聚合物骨架上共价接枝苯酚基团合成的,更便捷的合成策略仍具有挑战性。在此,我们开发了一种通过选择性正羟化聚(酪氨酸)合成多肽基多酚的通用而稳健的策略,通过金属-酚配位相互作用和 pH 可逆的苯硼酸酯键,分别构建了由 Cu2+ 和苯硼酸修饰的多柔比星原药(DOX-PBA)组成的动态纳米颗粒(GPCuD NPs)。此外,在聚乙二醇和多酚片段之间还加入了基质金属蛋白酶-2(MMP-2)可切除的 GPLGLAG 肽,通过酶触发的去乙酰化作用,使 GPCuD NPs 具有肿瘤特异性蓄积和深层渗透能力。值得注意的是,Cu2+螯合纳米粒子通过招募抗肿瘤免疫细胞和将M2型肿瘤相关巨噬细胞重新极化为M1表型,有效地改善了免疫抑制性肿瘤微环境。杯突酶驱动的金属免疫疗法与DOX化疗相结合,显著抑制了81.9%的4T1原位乳腺肿瘤生长,并建立了长期免疫记忆(有效记忆T细胞达30.8%),防止了肺转移。这项研究证明了一种可用于肿瘤靶向和杯突调节联合癌症金属免疫疗法的通用多酚纳米平台。
{"title":"Dynamic polyphenol nanoparticles boost cuproptosis-driven metalloimmunotherapy of breast cancer","authors":"","doi":"10.1016/j.nantod.2024.102442","DOIUrl":"10.1016/j.nantod.2024.102442","url":null,"abstract":"<div><p>Polyphenolic materials have been extensively explored for biomedical applications. However, most polymer-based polyphenols are synthesized by covalently grafting phenol groups on the backbone of polymers and more convenient synthetic strategy remains challenging. Herein, a general and robust strategy to synthesize polypeptide-based polyphenol via selective ortho-hydroxylation of poly(tyrosine) was developed to construct dynamic nanoparticles (GPCuD NPs) composed of Cu<sup>2+</sup> and phenylboronic acid modified doxorubicin prodrug (DOX-PBA) via metal-phenol coordination interaction and pH-reversible phenylboronate ester bond, respectively. Furthermore, matrix metalloproteinase-2 (MMP-2)-cleavable GPLGLAG peptide was also included between the polyethylene glycol and polyphenol segments, endowing GPCuD NPs with tumor-specific accumulation and deep penetration through enzyme-triggered depegylation. Notably, Cu<sup>2+</sup>-chelated nanoparticles efficiently ameliorated the immunosuppressive tumor microenvironment via recruiting antitumor immune cells and repolarizing M2-type tumor-associated macrophages to M1 phenotype. The combination of cuproptosis-driven metalloimmunotherapy with DOX chemotherapy remarkably suppressed 4T1 orthotopic breast tumor growth by 81.9 % and established a long-term immune memory (effective memory T cells up to 30.8 %) to prevent lung metastasis. This study has demonstrated a generalizable polyphenol nanoplatform for tumor-targeted and cuproptosis-regulated combination cancer metalloimmunotherapy.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141997465","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A multifunctional nutrient transfer nanoCRISPR scaffold induces metabolic remodeling to fuel cancer immunotherapy 多功能营养物质转移纳米CRISPR支架诱导新陈代谢重塑,为癌症免疫疗法提供动力
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-17 DOI: 10.1016/j.nantod.2024.102451

Tumor cells are major consumers of glutamine and glucose in the tumor microenvironment (TME), causing nutrient deficiency of immune cells, leading to immune escape and resistance to immunotherapy. However, pharmacological modulation would cause metabolic inhibition in both immune cells and tumor cells. Herein, a multifunctional nutrient transfer nanoCRISPR scaffold (FUEL) is fabricated to realize “nutrient transfer” from tumor cells to immune cells and remodel the metabolism in the TME, thus fueling cancer immunotherapy. FUEL is endowed with characteristics of enhanced blood circulation, specific tumor cell targeting, effective lysosomal escape, cascaded reactive-oxygen-species (ROS)-responsiveness, and ASCT2/GLUT1 dual gene knockout. Consequently, FUEL can restrict nutrient uptake of tumor cells thoroughly, increase glucose and glutamine in the TME remarkably to satisfy metabolic demands of immune cells, and reduce immunosuppressive metabolites concurrently. Metabolomics data shows that energy metabolism and biosynthesis are reduced in tumor cells but enhanced in immune cells. FUEL remarkably impedes the growth, metastasis, and recurrence of solid tumors in mice, further shows stronger anti-tumor immune responses and enhanced tumor inhibition in combination with anti-PD-L1 antibody. Overall, this nutrient transfer strategy enables a “one arrow aiming at three eagles” effect that induces a cascade amplification of antitumor immune responses for the maximized tumor therapy efficacy.

肿瘤细胞是肿瘤微环境(TME)中谷氨酰胺和葡萄糖的主要消耗者,造成免疫细胞营养缺乏,导致免疫逃逸和对免疫疗法的抵抗。然而,药物调节会导致免疫细胞和肿瘤细胞的代谢抑制。在此,我们制作了一种多功能营养转移纳米CRISPR支架(FUEL),以实现肿瘤细胞向免疫细胞的 "营养转移",重塑TME中的新陈代谢,从而为癌症免疫疗法提供助力。FUEL具有增强血液循环、特异性肿瘤细胞靶向、有效溶酶体逃逸、级联反应性氧物种(ROS)反应性和ASCT2/GLUT1双基因敲除等特点。因此,FUEL 能彻底限制肿瘤细胞对营养物质的摄取,显著增加 TME 中的葡萄糖和谷氨酰胺以满足免疫细胞的代谢需求,同时减少免疫抑制代谢物。代谢组学数据显示,肿瘤细胞的能量代谢和生物合成减少,而免疫细胞的能量代谢和生物合成增强。FUEL 显著抑制了实体瘤在小鼠体内的生长、转移和复发,与抗 PD-L1 抗体联用后,进一步显示出更强的抗肿瘤免疫反应和更强的肿瘤抑制能力。总之,这种营养物质转移策略能产生 "一箭三雕 "的效果,诱导抗肿瘤免疫反应的级联放大,从而实现肿瘤治疗效果的最大化。
{"title":"A multifunctional nutrient transfer nanoCRISPR scaffold induces metabolic remodeling to fuel cancer immunotherapy","authors":"","doi":"10.1016/j.nantod.2024.102451","DOIUrl":"10.1016/j.nantod.2024.102451","url":null,"abstract":"<div><p>Tumor cells are major consumers of glutamine and glucose in the tumor microenvironment (TME), causing nutrient deficiency of immune cells, leading to immune escape and resistance to immunotherapy. However, pharmacological modulation would cause metabolic inhibition in both immune cells and tumor cells. Herein, a multi<u>f</u>unctional n<u>u</u>trient transf<u>e</u>r nanoCRISPR scaffo<u>l</u>d (FUEL) is fabricated to realize “nutrient transfer” from tumor cells to immune cells and remodel the metabolism in the TME, thus fueling cancer immunotherapy. FUEL is endowed with characteristics of enhanced blood circulation, specific tumor cell targeting, effective lysosomal escape, cascaded reactive-oxygen-species (ROS)-responsiveness, and ASCT2/GLUT1 dual gene knockout. Consequently, FUEL can restrict nutrient uptake of tumor cells thoroughly, increase glucose and glutamine in the TME remarkably to satisfy metabolic demands of immune cells, and reduce immunosuppressive metabolites concurrently. Metabolomics data shows that energy metabolism and biosynthesis are reduced in tumor cells but enhanced in immune cells. FUEL remarkably impedes the growth, metastasis, and recurrence of solid tumors in mice, further shows stronger anti-tumor immune responses and enhanced tumor inhibition in combination with anti-PD-L1 antibody. Overall, this nutrient transfer strategy enables a “one arrow aiming at three eagles” effect that induces a cascade amplification of antitumor immune responses for the maximized tumor therapy efficacy.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141997463","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metal-based nanoparticles promote the activation of cGAS-STING pathway for enhanced cancer immunotherapy 金属基纳米粒子促进 cGAS-STING 通路的活化以增强癌症免疫疗法
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-17 DOI: 10.1016/j.nantod.2024.102445

Immunotherapy occupies an increasingly important place in the field of tumor treatment. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which is responsible for sensing and responding to cytosolic DNA, stands out as a key player of the host innate immunity and a significant contributor to anti-tumor immunotherapy. Besides, it serves as the principal signaling pathway for type I interferon (IFN) production, coordinating the maturation and activation of various immune cells like dendritic cells (DCs) and CD8+ T cells, thus bridging innate and adaptive immunity. The increasing focus on essential metal nanoparticles, notably Mn2+, Zn2+ and Ca2+, and their roles in the induction of oxidative stress are of increasing interest in the application of tumor immunotherapy especially for the stimulation of cGAS-STING pathway. Recent advancements in metal-based nanomaterials present a promising avenue for anti-tumor immunotherapy based on cGAS-STING pathway activation. This review offers a comprehensive overview of how metal-based nanomaterials affect the cGAS-STING pathway, as well as discusses the latest findings on metal-based nanomaterials, providing insights into their potential uses in cancer immunotherapy grounded in the activation of the cGAS-STING pathway.

免疫疗法在肿瘤治疗领域占据着越来越重要的地位。环GMP-AMP合成酶-干扰素基因刺激器(cGAS-STING)通路负责感知和响应细胞膜DNA,是宿主先天性免疫的关键角色,也是抗肿瘤免疫疗法的重要贡献者。此外,它还是 I 型干扰素(IFN)产生的主要信号通路,协调树突状细胞(DC)和 CD8+ T 细胞等各种免疫细胞的成熟和活化,从而连接先天性免疫和适应性免疫。人们越来越关注基本金属纳米粒子,特别是 Mn2+、Zn2+ 和 Ca2+,以及它们在诱导氧化应激方面的作用,这在肿瘤免疫疗法的应用中,特别是在刺激 cGAS-STING 通路方面,引起了越来越多的兴趣。金属基纳米材料的最新进展为基于 cGAS-STING 通路激活的抗肿瘤免疫疗法提供了一条前景广阔的途径。本综述全面概述了金属基纳米材料如何影响 cGAS-STING 通路,并讨论了有关金属基纳米材料的最新研究成果,深入探讨了它们在激活 cGAS-STING 通路的基础上用于癌症免疫疗法的潜力。
{"title":"Metal-based nanoparticles promote the activation of cGAS-STING pathway for enhanced cancer immunotherapy","authors":"","doi":"10.1016/j.nantod.2024.102445","DOIUrl":"10.1016/j.nantod.2024.102445","url":null,"abstract":"<div><p>Immunotherapy occupies an increasingly important place in the field of tumor treatment. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which is responsible for sensing and responding to cytosolic DNA, stands out as a key player of the host innate immunity and a significant contributor to anti-tumor immunotherapy. Besides, it serves as the principal signaling pathway for type I interferon (IFN) production, coordinating the maturation and activation of various immune cells like dendritic cells (DCs) and CD8<sup>+</sup> T cells, thus bridging innate and adaptive immunity. The increasing focus on essential metal nanoparticles, notably Mn<sup>2+</sup>, Zn<sup>2+</sup> and Ca<sup>2+</sup>, and their roles in the induction of oxidative stress are of increasing interest in the application of tumor immunotherapy especially for the stimulation of cGAS-STING pathway. Recent advancements in metal-based nanomaterials present a promising avenue for anti-tumor immunotherapy based on cGAS-STING pathway activation. This review offers a comprehensive overview of how metal-based nanomaterials affect the cGAS-STING pathway, as well as discusses the latest findings on metal-based nanomaterials, providing insights into their potential uses in cancer immunotherapy grounded in the activation of the cGAS-STING pathway.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141997467","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bioinspired Hf-based metal-organic framework radiosensitizer for nitric oxide-assisted radio-immunotherapy 用于一氧化氮辅助放射免疫疗法的生物启发铪基金属有机框架放射增敏剂
IF 13.2 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2024-08-17 DOI: 10.1016/j.nantod.2024.102447

Radiotherapy is a crucial antineoplastic approach in clinical practice, but it often suffers from low therapeutic efficacy due to inadequate deposition of X-ray radiation and limited inhibition of metastatic tumors. Here, a bioinspired Hf-based metal-organic framework (Hf-MOF) nano-radiosensitizer (SHMR) is elaborately designed for boosting radio-immunotherapy by synergizing radio-sensitization with nitric oxide-assisted immune microenvironment remodeling. The engineered SHMR is constructed by wrapping RGD peptide-modified erythrocyte membrane onto sodium nitroprusside-loaded Hf-MOF. In vitro and in vivo experiments demonstrate that SHMR can induce immunogenic cell death and release abundant tumor-associated antigens to promote dendritic cells maturation and T cells activation under X-ray irradiation. Importantly, nitric oxide (NO) released from SHMR can not only relieve tumor hypoxia to alleviate radiotherapy resistance, but also reprogram tumor microenvironment, thereby reshaping the extracellular matrix barrier and enhancing immune cells infiltration. Specifically, SHMR in conjunction with αPD-L1 therapy exhibits favorable therapeutic outcomes in bilateral tumor and metastatic tumor models. This work creates a practical nano-radiosensitizer to achieve effective radiotherapy and NO-mediated tumor microenvironment reconstruction, providing a promising strategy for potentiating the radio-immunotherapy against “immune-cold” tumors.

放射治疗是临床实践中一种重要的抗肿瘤方法,但由于 X 射线辐射沉积不足和对转移性肿瘤的抑制作用有限,其疗效往往较低。本文精心设计了一种生物启发的铪基金属有机框架(Hf-MOF)纳米放射增敏剂(SHMR),通过放射增敏与一氧化氮辅助免疫微环境重塑的协同作用来促进放射免疫治疗。这种工程化的 SHMR 是通过将 RGD 肽修饰的红细胞膜包裹在硝普钠负载的 Hf-MOF 上而构建的。体外和体内实验证明,在 X 射线照射下,SHMR 可诱导免疫原性细胞死亡,并释放丰富的肿瘤相关抗原,促进树突状细胞成熟和 T 细胞活化。重要的是,SHMR 释放的一氧化氮(NO)不仅能缓解肿瘤缺氧,减轻放疗耐药性,还能重塑肿瘤微环境,从而重塑细胞外基质屏障,增强免疫细胞浸润。具体来说,SHMR与αPD-L1疗法相结合,在双侧肿瘤和转移性肿瘤模型中显示出良好的治疗效果。这项工作创造了一种实用的纳米放射增敏剂,以实现有效的放射治疗和NO介导的肿瘤微环境重建,为放射免疫疗法对抗 "免疫冷 "肿瘤提供了一种前景广阔的策略。
{"title":"Bioinspired Hf-based metal-organic framework radiosensitizer for nitric oxide-assisted radio-immunotherapy","authors":"","doi":"10.1016/j.nantod.2024.102447","DOIUrl":"10.1016/j.nantod.2024.102447","url":null,"abstract":"<div><p>Radiotherapy is a crucial antineoplastic approach in clinical practice, but it often suffers from low therapeutic efficacy due to inadequate deposition of X-ray radiation and limited inhibition of metastatic tumors. Here, a bioinspired Hf-based metal-organic framework (Hf-MOF) nano-radiosensitizer (SHMR) is elaborately designed for boosting radio-immunotherapy by synergizing radio-sensitization with nitric oxide-assisted immune microenvironment remodeling. The engineered SHMR is constructed by wrapping RGD peptide-modified erythrocyte membrane onto sodium nitroprusside-loaded Hf-MOF. In vitro and in vivo experiments demonstrate that SHMR can induce immunogenic cell death and release abundant tumor-associated antigens to promote dendritic cells maturation and T cells activation under X-ray irradiation. Importantly, nitric oxide (NO) released from SHMR can not only relieve tumor hypoxia to alleviate radiotherapy resistance, but also reprogram tumor microenvironment, thereby reshaping the extracellular matrix barrier and enhancing immune cells infiltration. Specifically, SHMR in conjunction with αPD-L1 therapy exhibits favorable therapeutic outcomes in bilateral tumor and metastatic tumor models. This work creates a practical nano-radiosensitizer to achieve effective radiotherapy and NO-mediated tumor microenvironment reconstruction, providing a promising strategy for potentiating the radio-immunotherapy against “immune-cold” tumors.</p></div>","PeriodicalId":395,"journal":{"name":"Nano Today","volume":null,"pages":null},"PeriodicalIF":13.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141997466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Nano Today
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1