Photothermal ablation

光热烧蚀
  • 文章类型: Journal Article
    骨肉瘤是年轻人中最可怕的骨肿瘤之一。需要开发能够有效消除肿瘤的创新疗法,同时最大限度地减少对肢体功能的损害。理想的治疗策略应该具备三个基本能力:抗肿瘤作用,组织保护性能,和增强成骨能力。在这项研究中,自组装Ce取代的钼蓝(CMB)纳米轮晶体被合成并加载到3D打印的生物活性玻璃(CMB@BG)支架上,以开发一种独特的三合一骨肉瘤治疗方法。由于CMB纳米轮晶体的表面等离子体共振特性,CMB@BG支架在近红外II窗口内表现出出色的光热衍生肿瘤消融。此外,CMB的光热协同催化作用促进了过热引起的活性氧的快速清除,从而抑制炎症和保护周围组织。CMB@BG支架具有促进增殖和促进分化的能力,其有效地加速骨缺损内的骨再生。总之,结合高效肿瘤消融的CMB@BG支架,基于抗炎机制的组织保护,和增强成骨能力可能是解决骨肉瘤综合治疗需求的点对点解决方案。
    Osteosarcoma is one of the most dreadful bone neoplasms in young people, necessitating the development of innovative therapies that can effectively eliminate tumors while minimizing damage to limb function. An ideal therapeutic strategy should possess three essential capabilities: antitumor effects, tissue-protective properties, and the ability to enhance osteogenesis. In this study, self-assembled Ce-substituted molybdenum blue (CMB) nanowheel crystals are synthesized and loaded onto 3D-printed bioactive glass (CMB@BG) scaffolds to develop a unique three-in-one treatment approach for osteosarcoma. The CMB@BG scaffolds exhibit outstanding photothermally derived tumor ablation within the near-infrared-II window due to the surface plasmon resonance properties of the CMB nanowheel crystals. Furthermore, the photothermally synergistic catalytic effect of CMB promotes the rapid scavenging of reactive oxygen species caused by excessive heat, thereby suppressing inflammation and protecting surrounding tissues. The CMB@BG scaffolds possess pro-proliferation and pro-differentiation capabilities that efficiently accelerate bone regeneration within bone defects. Altogether, the CMB@BG scaffolds that combine highly efficient tumor ablation, tissue protection based on anti-inflammatory mechanisms, and enhanced osteogenic ability are likely to be a point-to-point solution for the comprehensive therapeutic needs of 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
    肿瘤免疫疗法已成为癌症治疗领域的有希望的前沿。然而,在实现本地化方面仍然存在挑战,持久的免疫刺激,同时抵消肿瘤的免疫抑制环境。这里,我们开发了一种基于天然贻贝足蛋白的纳米药物,具有时空控制,用于肿瘤免疫治疗。在这种纳米医学中,整合了免疫佐剂前药和光敏剂,这是由它们与蛋白质载体的动态键合和非共价组装驱动的。利用蛋白质载体的生物粘附,这种纳米药物实现了时空精确的药物共同递送,它不仅促进了肿瘤的光热消融,而且拓宽了肿瘤抗原库,促进具有持久性和维护性的原位免疫疗法。这种纳米药物还调节肿瘤微环境以克服免疫抑制,从而增强抗肿瘤应答对抗肿瘤进展。我们的策略强调了贻贝足蛋白衍生的药物递送设计理念,旨在完善组合免疫疗法,提供利用天然蛋白质治疗癌症的见解。
    Tumor immunotherapies have emerged as a promising frontier in the realm of cancer treatment. However, challenges persist in achieving localized, durable immunostimulation while counteracting the tumor\'s immunosuppressive environment. Here, we develop a natural mussel foot protein-based nanomedicine with spatiotemporal control for tumor immunotherapy. In this nanomedicine, an immunoadjuvant prodrug and a photosensitizer are integrated, which is driven by their dynamic bonding and non-covalent assembling with the protein carrier. Harnessing the protein carrier\'s bioadhesion, this nanomedicine achieves a drug co-delivery with spatiotemporal precision, by which it not only promotes tumor photothermal ablation but also broadens tumor antigen repertoire, facilitating in situ immunotherapy with durability and maintenance. This nanomedicine also modulates the tumor microenvironment to overcome immunosuppression, thereby amplifying antitumor responses against tumor progression. Our strategy underscores a mussel foot protein-derived design philosophy of drug delivery aimed at refining combinatorial immunotherapy, offering insights into leveraging natural proteins for cancer 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
    光热疗法(PTT)是一种通过使用光热材料和光敏剂来根除肿瘤组织的方法,这些光热材料和光敏剂从激光源中吸收光能并将其转化为热量,它选择性地靶向和破坏癌细胞,同时保留健康组织。已经深入研究了MXene作为PTT的光敏剂。然而,实现MXenes对肿瘤细胞的选择性仍然是一个挑战。针对肿瘤抗原的特异性抗体(Ab)可以实现光敏剂向肿瘤细胞的归巢,但是他们在MXene上的固定很少受到关注。这里,我们提供了使用MXene-聚多巴胺-抗CEACAM1Ab复合物选择性消融黑色素瘤细胞的策略。我们用聚多巴胺(PDA)涂覆Ti3C2TxMXene,一种将Ab附着在MXene表面的天然化合物,然后与抗CEACAM1Ab缀合。我们的实验证实了Ti3C2Tx-PDA和Ti3C2Tx-PDA-抗CEACAM1Ab复合物在各种细胞类型中的生物相容性。我们还建立了使用近红外辐射选择性消融CEACAM1阳性黑色素瘤细胞的方案。所获得的复合物在靶向和消除CEACAM1阳性黑素瘤细胞中表现出高选择性和效率,同时保留CEACAM1阴性细胞。这些结果证明了MXene-PDA-Ab复合物用于癌症治疗的潜力。他们强调了靶向治疗在肿瘤学中的关键作用,提供了一个有希望的途径,为精确和安全的治疗黑色素瘤和可能的其他以特定生物标志物为特征的癌症。未来的研究将旨在改善这些复合物的临床应用,为癌症治疗的新策略铺平道路。
    Photothermal therapy (PTT) is a method for eradicating tumor tissues through the use of photothermal materials and photosensitizing agents that absorb light energy from laser sources and convert it into heat, which selectively targets and destroys cancer cells while sparing healthy tissue. MXenes have been intensively investigated as photosensitizing agents for PTT. However, achieving the selectivity of MXenes to the tumor cells remains a challenge. Specific antibodies (Ab) against tumor antigens can achieve homing of the photosensitizing agents toward tumor cells, but their immobilization on MXene received little attention. Here, we offer a strategy for the selective ablation of melanoma cells using MXene-polydopamine-antiCEACAM1 Ab complexes. We coated Ti3C2Tx MXene with polydopamine (PDA), a natural compound that attaches Ab to the MXene surface, followed by conjugation with an anti-CEACAM1 Ab. Our experiments confirm the biocompatibility of the Ti3C2Tx-PDA and Ti3C2Tx-PDA-antiCEACAM1 Ab complexes across various cell types. We also established a protocol for the selective ablation of CEACAM1-positive melanoma cells using near-infrared irradiation. The obtained complexes exhibit high selectivity and efficiency in targeting and eliminating CEACAM1-positive melanoma cells while sparing CEACAM1-negative cells. These results demonstrate the potential of MXene-PDA-Ab complexes for cancer therapy. They underline the critical role of targeted therapies in oncology, offering a promising avenue for the precise and safe treatment of melanoma and possibly other cancers characterized by specific biomarkers. Future research will aim to refine these complexes for clinical use, paving the way for new strategies for 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
    与病原菌的发生密切相关,口腔鳞状细胞癌(OSCC)的发展和转移。抗菌治疗被认为是抑制细菌相关肿瘤和促进抗肿瘤免疫反应的增强策略。在这里,我们开发了一种可注射的粘合剂水凝胶,PNIPAM/DL@TIR,用于原位光热消融和针对牙龈卟啉单胞菌(Pg)定植的OSCC的抗肿瘤免疫的强大刺激,口腔主要致病菌之一。PNIPAM/DL@TIR,由聚(N-异丙基丙烯酰胺)组成,去甲基化木质素,和TAT肽缀合的IR820是使用简单的溶解-干燥-溶胀溶剂交换法制备的。在808nm激光照射下,PNIPAM/DL@TIR施加光热效应以消融Pg定植的OSCC并产生双重肿瘤和细菌抗原。由于其大量的儿茶酚基团,PNIPAM/DL@TIR有效地捕获这些抗原以形成原位抗原库,从而引发强大而持久的抗肿瘤免疫反应。蛋白质组学分析显示捕获的抗原包含肿瘤新抗原和细菌抗原。邻苯二酚基团赋予PNIPAM/DL@TIR抗氧化活性,这也有利于刺激抗肿瘤免疫力。总之,这项研究开发了一种可注射粘合剂水凝胶,并为治疗细菌相关性OSCC提供了一种组合策略.重要声明:在这项研究中,我们开发了一种可注射的粘合剂水凝胶,PNIPAM/DL@TIR,用于原位光热消融和针对牙龈卟啉单胞菌定植的OSCC的抗肿瘤免疫的强大刺激,口腔主要致病菌之一。PNIPAM/DL@TIR,由聚(N-异丙基丙烯酰胺)组成,去甲基化木质素,和TAT肽缀合的IR820表现出优异的光热性能。由于儿茶酚基团的存在,PNIPAM/DL@TIR具有良好的生物粘附特性,可以捕获蛋白质抗原以形成原位抗原库,从而启动强大和长期的抗肿瘤免疫反应。此外,PNIPAM/DL@TIR表现出较强的抗氧化活性,有利于提高抗肿瘤免疫力。在细菌感染的OSCC小鼠模型中,PNIPAM/DL@TIR不仅消融了NIR激光照射后的原发肿瘤,还可以诱导肿瘤和细菌原位接种以抑制远处肿瘤和肺转移。
    Pathogenic bacteria are closely associated with the occurrence, development and metastasis of oral squamous cell carcinoma (OSCC). Antibacterial therapy has been considered an enhancement strategy to suppress bacteria-associated tumors and promote anti-tumor immune responses. Herein, we developed an injectable adhesive hydrogel, PNIPAM/DL@TIR, for the in situ photothermal ablation and robust stimulation of antitumor immunity against OSCC colonized by Porphyromonas gingivalis (Pg), one of the major oral pathogenic bacteria. PNIPAM/DL@TIR, composed of poly(N-isopropylacrylamide), demethylated lignin, and TAT peptide-conjugated IR820, was prepared using a simple dissolve-dry-swell solvent exchange method. Upon 808 nm laser irradiation, PNIPAM/DL@TIR exerted photothermal effects to ablate Pg-colonized OSCC and generate dual tumor and bacterial antigens. Owing to its large number of catechol groups, PNIPAM/DL@TIR efficiently captured these antigens to form an in situ antigen repository, thereby eliciting robust and durable antitumor immune responses. Proteomic analysis revealed that the captured antigens comprised both tumor neoantigens and bacterial antigens. The catechol groups endowed PNIPAM/DL@TIR with antioxidant activity, which was also conducive to stimulating antitumor immunity. Altogether, this study develops an injectable adhesive hydrogel and provides a combination strategy for treating bacteria-associated OSCC. STATEMENT OF SIGNIFICANCE: In this study, we developed an injectable adhesive hydrogel, PNIPAM/DL@TIR, for in situ photothermal ablation and robust stimulation of antitumor immunity against OSCC colonized by Porphyromonas gingivalis, one of the major oral pathogenic bacteria. PNIPAM/DL@TIR, which consists of poly(N-isopropylacrylamide), demethylated lignin, and TAT peptide-conjugated IR820 exhibited outstanding photothermal performance. Owing to the presence of catechol groups, PNIPAM/DL@TIR has good bioadhesive properties and can capture protein antigens to form in situ antigen repository, thus initiating robust and long-term antitumor immune responses. In addition, PNIPAM/DL@TIR exhibited strong antioxidant activity that is favorable for promoting antitumor immunity. In the mouse model of OSCC with bacterial infection, PNIPAM/DL@TIR not only ablated the primary tumors upon NIR laser irradiation, but also induced tumor and bacterial vaccination in situ to suppress distant tumors and lung metastasis.
    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
    背景:早期乳腺癌(BC)迫切需要创新的治疗策略。源自乳腺导管细胞的肿瘤提供了靶向干预的机会。方法:我们探索了通过自然乳头开口进行导管内治疗作为早期BC的一种有希望的非侵入性方法。使用功能性近红外II(NIR-II)纳米材料,特别是NIR-IIb量子点与Epep多肽缀合,用于导管细胞靶向,我们对乳腺导管进行了原位成像和光热消融。导管内给药之后用808nm激光进行刺激。结果:该方法在微环境中实现了精确的导管破坏和增强的免疫反应。该技术在三阴性BC小鼠模型和导管原位癌大鼠模型中得到了验证,证明了局部BC治疗和预防的有希望的治疗潜力。结论:我们的研究证明了NIR-II纳米探针在引导乳腺导管的非侵入性光热消融中的有效性,为早期BC治疗提供了一条引人注目的途径。
    Background: Innovative treatment strategies for early-stage breast cancer (BC) are urgently needed. Tumors originating from mammary ductal cells present an opportunity for targeted intervention. Methods: We explored intraductal therapy via natural nipple openings as a promising non-invasive approach for early BC. Using functional Near-infrared II (NIR-II) nanomaterials, specifically NIR-IIb quantum dots conjugated with Epep polypeptide for ductal cell targeting, we conducted in situ imaging and photothermal ablation of mammary ducts. Intraductal administration was followed by stimulation with an 808 nm laser. Results: This method achieved precise ductal destruction and heightened immunological responses in the microenvironment. The technique was validated in mouse models of triple-negative BC and a rat model of ductal carcinoma in situ, demonstrating promising therapeutic potential for localized BC treatment and prevention. Conclusion: Our study demonstrated the effectiveness of NIR-II nanoprobes in guiding non-invasive photothermal ablation of mammary ducts, offering a compelling avenue for early-stage BC therapy.
    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
    聚集诱导发光剂(AIEgen)-功能化的有机-无机杂化纳米粒子(OINPs)是一类新兴的多功能纳米材料,具有广阔的潜在应用。AIEgen和无机化合物在AIEgen功能化OINPs中的空间排列和定位决定了结构,属性,和自组装纳米材料的功能。在这项工作中,我们提出了一种简便且通用的乳液自组装策略,用于通过将烷烃链官能化的无机纳米粒子与疏水性有机AIEgens共组装来合成定义明确的AIEgen官能化的OINP。作为概念的证明,通过使用烷烃链修饰的AuNP和AIEgens作为构建块,我们证明了等离子体-荧光混合纳米粒子(PFNP)从同心圆到核壳,然后到Janus结构的自组装和结构演化。通过改变AuNP表面上的烷烃配体长度和密度来控制AuNP在信号纳米复合材料中的空间位置。通过实验和理论模拟,还阐明了各种PFNP纳米结构形成的机理。获得的具有不同结构的PFNP具有空间可调的光学和光热特性,可用于多色和多模免疫标记以及光热灭菌的高级应用。这项工作提出了一种创新的合成方法,可以构建具有不同结构的AIEgen功能化OINP,Compositions,和功能,从而支持这些OINP的逐步发展。本文受版权保护。保留所有权利。
    Aggregation-induced emission luminogen (AIEgen)-functionalized organic-inorganic hybrid nanoparticles (OINPs) are an emerging category of multifunctional nanomaterials with vast potential applications. The spatial arrangement and positioning of AIEgens and inorganic compounds in AIEgen-functionalized OINPs determine the structures, properties, and functionalities of the self-assembled nanomaterials. In this work, a facile and general emulsion self-assembly tactic for synthesizing well-defined AIEgen-functionalized OINPs is proposed by coassembling alkane chain-functionalized inorganic nanoparticles with hydrophobic organic AIEgens. As a proof of concept, the self-assembly and structural evolution of plasmonic-fluorescent hybrid nanoparticles (PFNPs) from concentric circle to core shell and then to Janus structures is demonstrated by using alkane chain-modified AuNPs and AIEgens as building blocks. The spatial position of AuNPs in the signal nanocomposite is controlled by varying the alkane ligand length and density on the AuNP surface. The mechanism behind the formation of various PFNP nanostructures is also elucidated through experiments and theoretical simulation. The obtained PFNPs with diverse structures exhibit spatially tunable optical and photothermal properties for advanced applications in multicolor and multimode immunolabeling and photothermal sterilization. This work presents an innovative synthetic approach of constructing AIEgen-functionalized OINPs with diverse structures, compositions, and functionalities, thereby championing the progressive development of these OINPs.
    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
    治疗癌症的多学科方法似乎对于带来多个学科的益处及其在肿瘤消除中的协调至关重要。由于癌细胞的生物学和恶性特征,它们具有对常规疗法如化学疗法和放射疗法产生抗性的能力。胰腺癌(PC)是一种胃肠道恶性疾病,化疗和放疗是其治疗的主要手段。最近,纳米载体已成为其治疗中具有前景的结构。生物响应性纳米载体能够响应pH和氧化还原,其中,在有针对性的交付货物中,对PC进行特定的处理。纳米颗粒上的药物负载可以是合成或天然化合物,可以通过增强其在癌细胞中的细胞内积累来帮助减少PC的进展。纳米颗粒中基因的封装可以防止降解并促进肿瘤抑制中的细胞内积累。一种新的癌症疗法是光疗,其中纳米颗粒可以通过热疗和ROS过度生成刺激光热疗法和光动力疗法,从而触发PC中的细胞死亡。因此,在加速肿瘤抑制中进行光疗与化疗的协同治疗。纳米技术的重要功能之一是选择性靶向PC细胞以减少对正常细胞的副作用。纳米结构能够用适体进行表面官能化,蛋白质和抗体特异性靶向PC细胞以抑制其进展。因此,提供了一种针对PC的特异性疗法,并提出了对PC诊断的未来意义.
    The multidisciplinary approaches in treatment of cancer appear to be essential in term of bringing benefits of several disciplines and their coordination in tumor elimination. Because of the biological and malignant features of cancer cells, they have ability of developing resistance to conventional therapies such as chemo- and radio-therapy. Pancreatic cancer (PC) is a malignant disease of gastrointestinal tract in which chemotherapy and radiotherapy are main tools in its treatment, and recently, nanocarriers have been emerged as promising structures in its therapy. The bioresponsive nanocarriers are able to respond to pH and redox, among others, in targeted delivery of cargo for specific treatment of PC. The loading drugs on the nanoparticles that can be synthetic or natural compounds, can help in more reduction in progression of PC through enhancing their intracellular accumulation in cancer cells. The encapsulation of genes in the nanoparticles can protect against degradation and promotes intracellular accumulation in tumor suppression. A new kind of therapy for cancer is phototherapy in which nanoparticles can stimulate both photothermal therapy and photodynamic therapy through hyperthermia and ROS overgeneration to trigger cell death in PC. Therefore, synergistic therapy of phototherapy with chemotherapy is performed in accelerating tumor suppression. One of the important functions of nanotechnology is selective targeting of PC cells in reducing side effects on normal cells. The nanostructures are capable of being surface functionalized with aptamers, proteins and antibodies to specifically target PC cells in suppressing their progression. Therefore, a specific therapy for PC is provided and future implications for diagnosis of PC is suggested.
    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
    目前的抗癌研究表明,多种治疗方法的结合可以大大提高肿瘤细胞的杀伤能力。使用最新的微流体旋流混合器技术,联合化疗和光热消融治疗,我们开发了多响应靶向抗肿瘤纳米颗粒(NPs),由尺寸在200nm以下的叶酸官能化明胶NPs和包封的CuSNPs制成,Fe3O4NPs,和姜黄素(Cur)。通过探索明胶的结构,调整其浓度和pH,微调微流体装置中的流体动力学,获得了平均粒径为90±7nm的明胶NP的最佳制备条件。在肺腺癌A549细胞(低水平的叶酸受体)和乳腺癌MCF-7细胞(高水平的叶酸受体)上证明了药物递送系统(DDS)的比较靶向性。叶酸有助于实现NP向MCF-7肿瘤细胞的靶向和准确递送。协同光热消融和姜黄素的抗癌活性是通过红外光照射(980nm),而Fe3O4被外部磁场引导靶向明胶NP并加速药物的摄取,从而有效地杀死肿瘤细胞。这项工作中描述的方法很简单,容易重复,并且具有很大的潜力,可以扩大工业生产和随后的临床使用。
    Current anticancer research shows that a combination of multiple treatment methods can greatly improve the killing of tumor cells. Using the latest microfluidic swirl mixer technology, combined with chemotherapy and photothermal-ablation therapy, we developed multiresponsive targeted antitumor nanoparticles (NPs) made of folate-functionalized gelatin NPs under 200 nm in size and with encapsulated CuS NPs, Fe3O4 NPs, and curcumin (Cur). By exploring gelatin\'s structure, adjusting its concentration and pH, and fine-tuning the fluid dynamics in the microfluidic device, the best preparation conditions were obtained for gelatin NPs with an average particle size of 90 ± 7 nm. The comparative targeting of the drug delivery system (DDS) was demonstrated on lung adenocarcinoma A549 cells (low level of folate receptors) and breast adenocarcinoma MCF-7 cells (high level of folate receptors). Folic acid helps achieve targeting and accurate delivery of NPs to the MCF-7 tumor cells. The synergistic photothermal ablation and curcumin\'s anticancer activity are achieved through infrared light irradiation (980 nm), while Fe3O4 is guided with an external magnetic field to target gelatin NPs and accelerate the uptake of drugs, thus efficiently killing tumor cells. The method described in this work is simple, easy to repeat, and has great potential to be scaled up for industrial production and subsequent clinical use.
    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
    基于石墨烯的纳米材料具有有效的抗菌活性,并且作为对抗病原微生物的积极装甲引起了研究人员的极大兴趣。全面了解这些纳米材料的抗菌活性对于制造高效抗菌纳米材料至关重要,这导致高效和增强的活动。这些材料由于其抗微生物活性而被用作针对各种病原微生物的纳米药物。本文综述了石墨烯及其类似物如氧化石墨烯的抗菌活性,还原氧化石墨烯以及金属,金属氧化物和聚合物复合材料。该综述着重于各种纳米生物因素对抗菌能力的影响。它还提供了对这些材料的抗菌性能的深入了解,并简要讨论了科学界所证明的活性差异。这样,该综述有望阐明石墨烯纳米医学的未来研究和发展。
    Graphene-based nanomaterials possess potent antibacterial activity and have engrossed immense interest among researchers as an active armour against pathogenic microbes. A comprehensive perception of the antibacterial activity of these nanomaterials is critical to the fabrication of highly effective antimicrobial nanomaterials, which results in highly efficient and enhanced activity. These materials owing to their antimicrobial activity are utilized as nanomedicine against various pathogenic microbes. The present article reviews the antimicrobial activity of graphene and its analogs such as graphene oxide, reduced graphene oxide as well as metal, metal oxide and polymeric composites. The review draws emphasis on the effect of various nano-bio factors on the antibacterial capability. It also provides an insight into the antibacterial properties of these materials along with a brief discussion on the discrepancies in their activities as evidenced by the scientific communities. In this way, the review is expected to shed light on future research and development in graphene-based nanomedicine.
    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
    新型高亲水性和生物相容性的铋纳米球,金纳米颗粒在外面生长(Bi@Au纳米acantho球,Bi@AuNASs)是通过简单的程序合成的,由于具有超高的光热转换效率(η=46.6%),因此被证明是一种有前途的光热剂。制备的Bi@AuNASs对人肺癌A549细胞具有良好的血液相容性和相当低的细胞毒性,以及由近红外激光诱导的有效光热消融(PTA)治疗。在808nm激光辐射下,肿瘤温度可以升高到25°C,足以杀死癌细胞。此外,抗癌药物盐酸阿霉素(DOX)成功负载在Bi@AuNASs中,负载含量高达16.78%,并在pH敏感的释放曲线下释放,一种有利于将DOX静脉内递送到癌细胞中进行化疗的特征。Bi元素的存在使Bi@AuNASs可以作为CT成像引导的肿瘤治疗的有利计算机断层扫描(CT)造影剂。与通过光热疗法或化学疗法治疗癌症相比,使用Bi@AuNASs作为光热剂和药物载体的化学-光热协同疗法有效地增强了癌症治疗中的体外和体内治疗效果。
    Novel highly hydrophilic and biocompatible bismuth nanospheres with gold nanoparticles growing outside (Bi@Au nano-acanthospheres, Bi@Au NASs) were synthesized through a simple procedure, which demonstrated to be a promising photothermal agent owing to the ultrahigh photothermal conversion efficiency (η = 46.6 %). The as-prepared Bi@Au NASs showed excellent blood compatibility and fairly low cytotoxicity to human lung cancer A549 cells, as well as efficient photothermal ablation (PTA) therapy induced by a near-infrared laser. Under the 808 nm laser radiation, the tumour temperature could be elevated by ∼25 °C high enough to kill the cancer cells. Moreover, the anticancer drug doxorubicin hydrochloride (DOX) was successfully loaded in Bi@Au NASs with a loading content as high as 16.78 % and released under a pH sensitive release profile, a characteristic beneficial for intravenous delivery of DOX into cancer cells for chemotherapy. The presence of the Bi element enabled Bi@Au NASs to act as a favourable computed tomography (CT) contrast medium for CT imaging-guided tumour treatment. Compared with cancer treatment through either photothermal therapy or chemotherapy, the chemo-photothermal synergistic therapy using Bi@Au NASs as both a photothermal agent and a drug carrier has efficiently enhanced the in vitro and in vivo therapeutic effects in cancer treatment.
    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