DLS, dynamic light scattering

DLS,动态光散射
  • 文章类型: Journal Article
    角膜移植术是临床治疗角膜疾病的有效方法,which,然而,受到供体角膜的限制。开发具有“透明”和“上皮和基质生成”功能的生物粘附性角膜补片具有重要的临床价值,以及“无情”和“坚韧”。同时满足\"T.E.S.T.“要求,基于甲基丙烯酰化明胶(GelMA)设计了一种光固化水凝胶,PluronicF127二丙烯酸酯(F127DA)和醛化PluronicF127(AF127)共组装双功能胶束和I型胶原蛋白(COLI),结合临床应用的角膜交联(CXL)技术修复受损角膜。紫外线照射5分钟后形成的贴片具有透明,非常艰难,和强大的生物粘合性能。多次交联使贴片承受近600%的变形,并表现出大于400mmHg的爆裂压力,显著高于正常眼压(10-21mmHg)。此外,与无COLI的GelMA-F127DA和AF127水凝胶相比,降解速度较慢,使水凝胶贴片在体内基质床上稳定,支持角膜上皮和基质的再生。水凝胶贴剂可在4周内替代角膜深层基质缺损,并能很好地生物整合到兔模型的角膜组织中,联合CXL在圆锥角膜和其他角膜疾病的手术中显示出巨大的潜力。
    Corneal transplantation is an effective clinical treatment for corneal diseases, which, however, is limited by donor corneas. It is of great clinical value to develop bioadhesive corneal patches with functions of \"Transparency\" and \"Epithelium & Stroma generation\", as well as \"Suturelessness\" and \"Toughness\". To simultaneously meet the \"T.E.S.T.\" requirements, a light-curable hydrogel is designed based on methacryloylated gelatin (GelMA), Pluronic F127 diacrylate (F127DA) & Aldehyded Pluronic F127 (AF127) co-assembled bi-functional micelles and collagen type I (COL I), combined with clinically applied corneal cross-linking (CXL) technology for repairing damaged cornea. The patch formed after 5 min of ultraviolet irradiation possesses transparent, highly tough, and strongly bio-adhesive performance. Multiple cross-linking makes the patch withstand deformation near 600% and exhibit a burst pressure larger than 400 mmHg, significantly higher than normal intraocular pressure (10-21 mmHg). Besides, the slower degradation than GelMA-F127DA&AF127 hydrogel without COL I makes hydrogel patch stable on stromal beds in vivo, supporting the regrowth of corneal epithelium and stroma. The hydrogel patch can replace deep corneal stromal defects and well bio-integrate into the corneal tissue in rabbit models within 4 weeks, showing great potential in surgeries for keratoconus and other corneal diseases by combining with CXL.
    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
    纳米颗粒表面上蛋白质冠的存在调节它们的生理相互作用,例如细胞缔合和靶向性质。已经显示,负载α-甘露聚糖(αM)的聚(乙二醇)-聚(1-丙交酯)(PEG-PLA)纳米颗粒(NP-αM)特异性地增加小胶质细胞中低密度脂蛋白受体(LDLR)的表达,并改善多次施用后淀粉样蛋白β(Aβ)的清除率。然而,纳米粒子如何穿过血脑屏障和进入小胶质细胞仍然是未知的。这里,我们研究了PEG-PLA纳米颗粒在不同条件下的脑递送性能,发现纳米颗粒在αM加载和多次给药后表现出更高的脑转运效率和小胶质细胞摄取效率。为了揭示机制,我们进行了蛋白质组学分析,以表征在各种条件下形成的蛋白质冠的组成,发现药物负载和多次给药都会影响蛋白质冠的组成,并随后影响b.End3和BV-2细胞中纳米颗粒的细胞摄取。补体蛋白,免疫球蛋白,发现RAB5A和CD36在电晕中富集,并与纳米颗粒的摄取过程有关。总的来说,我们带来了关于蛋白质电晕在靶向药物递送中的调节作用的机械理解,并为工程化脑或小胶质细胞特异性靶向给药系统提供理论依据。
    The presence of protein corona on the surface of nanoparticles modulates their physiological interactions such as cellular association and targeting property. It has been shown that α-mangostin (αM)-loaded poly(ethylene glycol)-poly(l-lactide) (PEG-PLA) nanoparticles (NP-αM) specifically increased low density lipoprotein receptor (LDLR) expression in microglia and improved clearance of amyloid beta (Aβ) after multiple administration. However, how do the nanoparticles cross the blood‒brain barrier and access microglia remain unknown. Here, we studied the brain delivery property of PEG-PLA nanoparticles under different conditions, finding that the nanoparticles exhibited higher brain transport efficiency and microglia uptake efficiency after αM loading and multiple administration. To reveal the mechanism, we performed proteomic analysis to characterize the composition of protein corona formed under various conditions, finding that both drug loading and multiple dosing affect the composition of protein corona and subsequently influence the cellular uptake of nanoparticles in b.End3 and BV-2 cells. Complement proteins, immunoglobulins, RAB5A and CD36 were found to be enriched in the corona and associated with the process of nanoparticles uptake. Collectively, we bring a mechanistic understanding about the modulator role of protein corona on targeted drug delivery, and provide theoretical basis for engineering brain or microglia-specific targeted delivery system.
    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
    光热疗法具有微创的特点,可控性,效率高,特异性强,能有效弥补传统药物治疗带来的毒副作用和肿瘤耐药性。然而,由于红外光的组织穿透力有限,很难在临床上推广应用。眼睛是人类唯一的透明组织,红外光可以很容易地穿透眼睛组织,因此光热疗法有望用于治疗眼底疾病。在这里,由脂质体和吲哚菁绿(ICG)组装而成的新型纳米平台用于治疗视网膜母细胞瘤.将ICG组装在脂质体中以克服ICG本身的一些问题。例如,ICG很容易淬火,自我聚集和不稳定。此外,脂质体可以防止游离ICG通过体循环被清除。纳米平台的构建不仅保证了ICG在体内的稳定性,而且还实现了成像引导光热治疗,这创造了一种治疗视网膜母细胞瘤的新策略。
    Photothermal therapy has the characteristics of minimal invasiveness, controllability, high efficiency, and strong specificity, which can effectively make up for the toxic side effects and tumor resistance caused by traditional drug treatment. However, due to the limited tissue penetration of infrared light, it is difficult to promote and apply in clinical practice. The eye is the only transparent tissue in human, and infrared light can easily penetrate the eye tissue, so it is expected that photothermal therapy can be used to treat fundus diseases. Here in, a new nano-platform assembled by liposome and indocyanine green (ICG) was used to treat retinoblastoma. ICG was assembled in liposomes to overcome some problems of ICG itself. For example, ICG is easily quenched, self-aggregating and instability. Moreover, liposomes can prevent free ICG from being cleared through the systemic circulation. The construction of the nano-platform not only ensured the stability of ICG in vivo, but also realized imaging-guide photothermal therapy, which created a new strategy for the treatment of retinoblastoma.
    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
    通过重编程肿瘤相关巨噬细胞(TAM)重塑肿瘤微环境和通过免疫原性细胞死亡(ICD)增加肿瘤的免疫原性已成为有前途的抗癌免疫治疗策略。然而,TAMs在肿瘤组织中的异质性分布和肿瘤细胞的异质性使得免疫激活具有挑战性。为了克服这些困境,一种具有肿瘤靶向和渗透的杂交细菌,TAM极化,和光热转化能力被开发用于改善体内抗肿瘤免疫疗法。杂种细菌(B.b@QDs)是通过将Ag2S量子点(QDs)负载在两歧双歧杆菌(B.b)通过静电相互作用。具有缺氧靶向能力的杂合菌可有效蓄积并穿透肿瘤组织,使B.b与TAM充分接触并介导它们向M1表型的极化,以逆转免疫抑制性肿瘤微环境。通过将B.b的肿瘤渗透与QDs的光热效应耦合,还可以克服肿瘤内异质性并获得丰富的肿瘤相关抗原,导致增强的免疫效果。这种结合了B.b触发的TAM极化和QD诱导的ICD的策略在原位乳腺癌中实现了对肿瘤生长的显着抑制。
    Remodeling the tumor microenvironment through reprogramming tumor-associated macrophages (TAMs) and increasing the immunogenicity of tumors via immunogenic cell death (ICD) have been emerging as promising anticancer immunotherapy strategies. However, the heterogeneous distribution of TAMs in tumor tissues and the heterogeneity of the tumor cells make the immune activation challenging. To overcome these dilemmas, a hybrid bacterium with tumor targeting and penetration, TAM polarization, and photothermal conversion capabilities is developed for improving antitumor immunotherapy in vivo. The hybrid bacteria (B.b@QDs) are prepared by loading Ag2S quantum dots (QDs) on the Bifidobacterium bifidum (B.b) through electrostatic interactions. The hybrid bacteria with hypoxia targeting ability can effectively accumulate and penetrate the tumor tissues, enabling the B.b to fully contact with the TAMs and mediate their polarization toward M1 phenotype to reverse the immunosuppressive tumor microenvironment. It also enables to overcome the intratumoral heterogeneity and obtain abundant tumor-associated antigens by coupling tumor penetration of the B.b with photothermal effect of the QDs, resulting in an enhanced immune effect. This strategy that combines B.b-triggered TAM polarization and QD-induced ICD achieved a remarkable inhibition of tumor growth in orthotopic breast 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
    中风是导致死亡和残疾的主要原因。目前,这种疾病没有有效的药物治疗方法,这可以部分归因于无法有效地向大脑提供治疗。在这里,我们报告了天然化合物衍生的纳米颗粒(NPs)的发展,它既可以作为中风治疗的有效治疗剂,又可以作为向缺血性大脑输送药物的有效载体。首先,我们筛选了一组天然纳米材料,并确定了桦木酸(BA)是治疗中风最有效的抗氧化剂之一.接下来,我们设计了BANP,通过将BA化学转化为桦木胺(BAM),在酸性缺血组织中优先释放药物,并通过CXCR4拮抗剂AMD3100的表面缀合进行靶向药物递送。所得的AMD3100-共轭BAMNP,或A-BAMNP,然后被评估为用于中风治疗的治疗剂和作为用于递送神经保护肽NA1的载体。我们表明,A-BAMNP的静脉给药有效地改善了中风的恢复,并且当NA1被封装时,其功效进一步增强。由于它们的多功能性和显著的功效,我们预计,A-BAMNP有可能同时作为治疗药物和药物载体来改善卒中的治疗.
    Stroke is the leading cause of death and disability. Currently, there is no effective pharmacological treatment for this disease, which can be partially attributed to the inability to efficiently deliver therapeutics to the brain. Here we report the development of natural compound-derived nanoparticles (NPs), which function both as a potent therapeutic agent for stroke treatment and as an efficient carrier for drug delivery to the ischemic brain. First, we screened a collection of natural nanomaterials and identified betulinic acid (BA) as one of the most potent antioxidants for stroke treatment. Next, we engineered BA NPs for preferential drug release in acidic ischemic tissue through chemically converting BA to betulinic amine (BAM) and for targeted drug delivery through surface conjugation of AMD3100, a CXCR4 antagonist. The resulting AMD3100-conjugated BAM NPs, or A-BAM NPs, were then assessed as a therapeutic agent for stroke treatment and as a carrier for delivery of NA1, a neuroprotective peptide. We show that intravenous administration of A-BAM NPs effectively improved recovery from stroke and its efficacy was further enhanced when NA1 was encapsulated. Due to their multifunctionality and significant efficacy, we anticipate that A-BAM NPs have the potential to be translated both as a therapeutic agent and as a drug carrier to improve the treatment of stroke.
    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
    尽管几种人工纳米疗法已被批准用于转移性乳腺癌的实际治疗,他们低效的治疗结果,严重的不良影响,大规模生产的高成本仍然是关键的挑战。在这里,我们开发了一种替代策略,通过使用来自茶花的天然纳米载体(TFEN)特异性触发乳腺肿瘤细胞凋亡并抑制其肺转移.这些纳米载体具有理想的粒径(131nm),外泌体样形态,和负zeta电位。此外,TFEN被发现含有大量的多酚,黄酮类化合物,功能蛋白,和脂质。细胞实验表明,由于刺激活性氧(ROS)扩增,TFEN对癌细胞显示出强细胞毒性。细胞内ROS数量的增加不仅可以触发线粒体损伤,但也阻止细胞周期,导致体外抗增殖,反移民,和抗乳腺癌细胞侵袭活性。进一步的小鼠研究表明,静脉内(i.v.)注射或口服给药后的TFEN可以在乳腺肿瘤和肺转移部位积聚,抑制乳腺癌的生长和转移,并调节肠道微生物群。这项研究为通过静脉内和口服途径抑制乳腺癌及其肺转移的天然外泌体样纳米平台的绿色生产带来了新的见解。
    Although several artificial nanotherapeutics have been approved for practical treatment of metastatic breast cancer, their inefficient therapeutic outcomes, serious adverse effects, and high cost of mass production remain crucial challenges. Herein, we developed an alternative strategy to specifically trigger apoptosis of breast tumors and inhibit their lung metastasis by using natural nanovehicles from tea flowers (TFENs). These nanovehicles had desirable particle sizes (131 nm), exosome-like morphology, and negative zeta potentials. Furthermore, TFENs were found to contain large amounts of polyphenols, flavonoids, functional proteins, and lipids. Cell experiments revealed that TFENs showed strong cytotoxicities against cancer cells due to the stimulation of reactive oxygen species (ROS) amplification. The increased intracellular ROS amounts could not only trigger mitochondrial damage, but also arrest cell cycle, resulting in the in vitro anti-proliferation, anti-migration, and anti-invasion activities against breast cancer cells. Further mice investigations demonstrated that TFENs after intravenous (i.v.) injection or oral administration could accumulate in breast tumors and lung metastatic sites, inhibit the growth and metastasis of breast cancer, and modulate gut microbiota. This study brings new insights to the green production of natural exosome-like nanoplatform for the inhibition of breast cancer and its lung metastasis via i.v. and oral routes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Ferroptosis因其在消除耐药肿瘤细胞方面无与伦比的机制而受到越来越多的关注。然而,毒性脂质过氧化物(LPO)在肿瘤部位的积累受到脂质氧化水平的限制。在这里,通过利用多功能海藻酸钠(ALG)水凝胶,由藤黄酸(GA)组成的局部铁中毒触发剂,2,2'-偶氮双[2-(2-咪唑啉-2-基)丙烷]二盐酸盐(AIPH),和墨水(光热剂),通过简单的瘤内注射构建。在1064​nm激光照射下,储存的AIPH迅速分解成烷基(R•),这加重了肿瘤细胞中的LPO。同时,GA能抑制热休克蛋白90(HSP90)降低肿瘤细胞的耐热性,并强制消耗谷胱甘肽(GSH)以削弱细胞的抗氧化能力。系统的体外和体内实验表明,GSH的同步消耗和活性氧(ROS)的增加促进了谷胱甘肽过氧化物酶4(GPX4)的表达降低,这进一步导致了细胞内氧化还原稳态的破坏,并最终促进了铁细胞凋亡。这种多合一策略通过在肿瘤部位消耗和产生致命的活性化合物而具有高度有效的肿瘤抑制作用,这将为可控的,准确,和协调的肿瘤治疗。
    Ferroptosis has received ever-increasing attention due to its unparalleled mechanism in eliminating resistant tumor cells. Nevertheless, the accumulation of toxic lipid peroxides (LPOs) at the tumor site is limited by the level of lipid oxidation. Herein, by leveraging versatile sodium alginate (ALG) hydrogel, a localized ferroptosis trigger consisting of gambogic acid (GA), 2,2\'-azobis [2-(2-imidazolin-2-yl) propane] dihydrochloride (AIPH), and Ink (a photothermal agent), was constructed via simple intratumor injection. Upon 1064 ​nm laser irradiation, the stored AIPH rapidly decomposed into alkyl radicals (R•), which aggravated LPOs in tumor cells. Meanwhile, GA could inhibit heat shock protein 90 (HSP90) to reduce the heat resistance of tumor cells, and forcefully consume glutathione (GSH) to weaken the antioxidant capacity of cells. Systematic in vitro and in vivo experiments have demonstrated that synchronous consumption of GSH and increased reactive oxygen species (ROS) facilitated reduced expression of glutathione peroxidase 4 (GPX4), which further contributed to disruption of intracellular redox homeostasis and ultimately boosted ferroptosis. This all-in-one strategy has a highly effective tumor suppression effect by depleting and generating fatal active compounds at tumor sites, which would pave a new route for the controllable, accurate, and coordinated tumor treatments.
    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
    肿瘤转移是化疗失败和癌症相关死亡的原因。此外,循环肿瘤细胞(CTC)簇在肿瘤转移中起关键作用。在这里,我们开发了癌症特异性钙纳米调节剂,通过癌症膜包被的地高辛(DIG)和多柔比星(DOX)共包裹的PLGA纳米颗粒(CPDD)来抑制CTC簇的产生和循环。CPDD可以精确靶向血液和淋巴循环中的同源原代肿瘤细胞和CTC簇。有趣的是,CPDDs通过抑制Na+/K+-ATP酶诱导细胞内Ca2+的积累,这有助于抑制细胞-细胞连接解聚CTC簇。同时,CPDD抑制上皮-间质转化(EMT)过程,导致抑制肿瘤细胞从原发部位逃逸。此外,DOX和DIG以5:1的质量比的组合协同诱导肿瘤细胞的凋亡。体内外实验结果表明,CPDDs不仅能有效抑制CTC簇的产生和循环,而且精确靶向和消除原发性肿瘤。我们的发现为抗转移联合化疗提供了一种新的方法。
    Tumor metastasis is responsible for chemotherapeutic failure and cancer-related death. Moreover, circulating tumor cell (CTC) clusters play a pivotal role in tumor metastasis. Herein, we develop cancer-specific calcium nanoregulators to suppress the generation and circulation of CTC clusters by cancer membrane-coated digoxin (DIG) and doxorubicin (DOX) co-encapsulated PLGA nanoparticles (CPDDs). CPDDs could precisely target the homologous primary tumor cells and CTC clusters in blood and lymphatic circulation. Intriguingly, CPDDs induce the accumulation of intracellular Ca2+ by inhibiting Na+/K+-ATPase, which help restrain cell-cell junctions to disaggregate CTC clusters. Meanwhile, CPDDs suppress the epithelial-mesenchymal transition (EMT) process, resulting in inhibiting tumor cells escape from the primary site. Moreover, the combination of DOX and DIG at a mass ratio of 5:1 synergistically induces the apoptosis of tumor cells. In vitro and in vivo results demonstrate that CPDDs not only effectively inhibit the generation and circulation of CTC clusters, but also precisely target and eliminate primary tumors. Our findings present a novel approach for anti-metastasis combinational chemotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    癌症治疗的主要挑战是如何有效消除原发性肿瘤并充分诱导免疫原性细胞死亡(ICD)以激发强大的免疫反应来控制转移。这里,开发了一种自组装的级联生物反应器,以增强肿瘤渗透和饥饿的协同治疗来改善癌症治疗,化学动力学(CDT)和光热疗法。以葡萄糖氧化酶(GOx)为模板合成超小FeS-GOx纳米点,紫杉醇(PTX)通过疏水作用诱导形成自组装FeS-GOx@PTX(FGP)。在肿瘤部位积累后,FGP分解为较小的FeS-GOx,以增强肿瘤的深层渗透。GOx维持高的酶活性以在氧的辅助下催化葡萄糖以产生过氧化氢(H2O2)作为饥饿疗法。涉及再生H2O2的Fenton反应进而产生更多的羟基自由基以增强CDT。跟随808nm的近红外激光,通过联合治疗,FGP在体外和体内显示出显著的肿瘤抑制。随之而来的钙网织蛋白暴露增加了ICD并促进了树突状细胞的成熟。结合抗CTLA4检查点封锁,由于细胞毒性T淋巴细胞的肿瘤内浸润增强,FGP可以绝对消除原发性肿瘤并积极抑制远处肿瘤。我们的工作提出了一种有希望的原发性肿瘤和转移抑制策略。
    Major challenges for cancer treatment are how to effectively eliminate primary tumor and sufficiently induce immunogenic cell death (ICD) to provoke a robust immune response for metastasis control. Here, a self-assembled cascade bioreactor was developed to improve cancer treatment with enhanced tumor penetration and synergistic therapy of starvation, chemodynamic (CDT) and photothermal therapy. Ultrasmall FeS-GOx nanodots were synthesized with glucose oxidase (GOx) as template and induced by paclitaxel (PTX) to form self-assembling FeS-GOx@PTX (FGP) via hydrophobic interaction. After accumulated at tumor sites, FGP disassembles to smaller FeS-GOx for enhanced deep tumor penetration. GOx maintains high enzymatic activity to catalyze glucose with assistant of oxygen to generate hydrogen peroxide (H2O2) as starvation therapy. Fenton reaction involving the regenerated H2O2 in turn produced more hydroxyl radicals for enhanced CDT. Following near-infrared laser at 808 nm, FGPs displayed pronounced tumor inhibition in vitro and in vivo by the combination therapy. The consequent increased exposure to calreticulin amplified ICD and promoted dendritic cells maturation. In combination with anti-CTLA4 checkpoint blockade, FGP can absolutely eliminate primary tumor and avidly inhibit distant tumors due to the enhanced intratumoral infiltration of cytotoxic T lymphocytes. Our work presents a promising strategy for primary tumor and metastasis inhibition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    肝细胞癌(HCC)已被称为全球第二常见的主要癌症,因为它对化疗和药物的反应都很差。雷公藤甲素(TP),二萜三环氧化物,是一种有前途的治疗剂,因为它对包括HCC在内的多种癌症具有有效的抗癌作用。然而,由于其严重的全身毒性,其临床应用受到限制,低溶解度,在体内快速消除。因此,为了克服上述障碍,设计了光敏剂Ce6和化疗药物TP(TP/Ce6-LP)整合的可光活化脂质体(LP),以追求HCC治疗中的药物控释和协同光动力疗法。由于增强的通透性和滞留(EPR)效应,包封在脂质体中的TP积累到肿瘤部位。在激光照射下,光敏剂Ce6产生活性氧(ROS)并进一步氧化不饱和磷脂。这样,脂质体被破坏以释放TP。用NIR激光照射的TP/Ce6-LP(TP/Ce6-LPL)在体外和体内对患者来源的HCC肿瘤异种移植物(PDXHCC)均显示出最佳的抗肿瘤作用。TP/Ce6-LP可显著降低TP的副作用。此外,TP/Ce6-LP+L通过caspase-3/PARP信号通路诱导细胞凋亡。总的来说,TP/Ce6-LP+L是一种新的潜在治疗选择,在停止肝癌进展与毒性减弱。
    Hepatocellular carcinoma (HCC) has been known as the second common leading cancer worldwide, as it responds poorly to both chemotherapy and medication. Triptolide (TP), a diterpenoid triepoxide, is a promising treatment agent for its effective anticancer effect on multiple cancers including HCC. However, its clinical application has been limited owing to its severe systemic toxicities, low solubility, and fast elimination in the body. Therefore, to overcome the above obstacles, photo-activatable liposomes (LP) integrated with both photosensitizer Ce6 and chemotherapeutic drug TP (TP/Ce6-LP) was designed in the pursuit of controlled drug release and synergetic photodynamic therapy in HCC therapy. The TP encapsulated in liposomes accumulated to the tumor site due to the enhanced permeability and retention (EPR) effect. Under laser irradiation, the photosensitizer Ce6 generated reactive oxygen species (ROS) and further oxidized the unsaturated phospholipids. In this way, the liposomes were destroyed to release TP. TP/Ce6-LP with NIR laser irradiation (TP/Ce6-LP+L) showed the best anti-tumor effect both in vitro and in vivo on a patient derived tumor xenograft of HCC (PDXHCC). TP/Ce6-LP significantly reduced the side effects of TP. Furthermore, TP/Ce6-LP+L induced apoptosis through a caspase-3/PARP signaling pathway. Overall, TP/Ce6-LP+L is a novel potential treatment option in halting HCC progression with attenuated toxicity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号