In vivo delivery of miRNAs for cancer therapy Challenges and strategies

合集下载

在干细胞药物临床前研究中动物样品基因组DNA高通量提取的固相萃取法和磁珠筛选法初探

在干细胞药物临床前研究中动物样品基因组DNA高通量提取的固相萃取法和磁珠筛选法初探

・2372・弘总铺硝觅Drug Evaluation Research第43卷第12期2020年12月在干细胞药物临床前研究中动物样品基因组DNA高通量提取的固相萃取法和磁珠筛选法初探张臣,谭锦绣,李薇°天津天诚新药评价有限公司国家释药技术及药代动力学重点实验室,天津300310摘要:干细胞药物临床前研究中,常需要以干细胞特定基因片段为靶点追踪其体内的分布过程,需要提取动物样品基因组DNA,涉及到心脏、肝脏、肺脏、脾脏、肾脏、脑、骨髓、血、脂肪、肌肉、性腺、胃和肠等多种组织类型。

为实现临床前样品的高通量检测,总结动物组织基因组DNA提取方法,并就如何高效高质提取样品基因组DNA进行讨论,提出间充质干细胞临床前研究中样品处理的一种可行的方案:通过生物样品制备仪获取稳定均一的组织裂解液,利用固相萃取法和磁珠筛选法配合使用获得高质量高浓度的动物样品DNAo关键词:DNA提取;固相萃取;磁珠筛选;干细胞中图分类号:R329文献标志码:A文章编号:1674・6376(2020)12-2372-03DOI:10.7501/j.issn.1674-6376.2020.12.002Preliminary study of high-throughput animal samples genomic DNA extraction scheme by solid phase extraction and magnetic bead screening in preclinical research of stem cell drugsZHANG Chen,TAN Jinxiu,LI WeiTianjin Tianchen New Drug Evaluation Research Co.LTD,State key laboratory of drug release technology and pharmacokinetics, Tianjin300031,ChinaAbstract:In preclinical research of stem cell drugs,it was often necessary to track the distribution process of stem cell specific gene fragments in vivo,and extract genomic DNA of animal samples,involving heart,liver,lung,spleen,kidney,brain,bone marrow, blood,fat,muscle,gonad,stomach and intestine.For such a special purpose of DNA extraction,there was no systematic report.In this paper,the methods of extracting genomic DNA from animal tissues were summarized,and how to extract genomic DNA from samples with high efficiency and high quality was discussed.A feasible scheme for sample treatment in preclinical research of mesenchymal stem cells was proposed:stable and uniform tissue lysate was obtained by biological sample preparation apparatus, and high quality and high concentration animal sample DNA was obtained by solid phase extraction and magnetic bead screening. Key words:DNA extraction;solid phase extraction;magnetic bead screening;stem cells利用定量PCR(qPCR)进行干细胞药物临床前评价分析时(⑷,从实验动物样品中稳定地提取高浓度、高质量的基因组DNA,对临床前研究的顺利完成至关重要。

替雷利珠单抗联合贝伐珠单抗治疗不可切除晚期肝癌的临床研究

替雷利珠单抗联合贝伐珠单抗治疗不可切除晚期肝癌的临床研究

- 41 -*基金项目:2021年阳江市科技局科研项目(SF2021016)①阳江市人民医院 广东 阳江 529500替雷利珠单抗联合贝伐珠单抗治疗不可切除晚期肝癌的临床研究*叶天允① 张启红① 许云朋①【摘要】 目的:探讨替雷利珠单抗联合贝伐珠单抗在不可切除晚期肝癌患者中的临床效果。

方法:选择2021年7月—2023年1月阳江市人民医院肿瘤一科收治的96例不可切除晚期肝癌患者作为研究对象,根据随机数表法分为两组,各48例。

对照组口服索拉菲尼治疗,观察组给予替雷利珠单抗联合贝伐珠单抗静脉滴注治疗。

比较两组近期疗效、生化指标[甲胎蛋白(AFP)、铁蛋白、γ-谷氨酰转肽酶(GGT)]及毒副反应发生率。

结果:治疗3个月后,观察组总有效率高于对照组,差异有统计学意义(P <0.05)。

治疗前,两组生化指标比较,差异无统计学意义(P >0.05);治疗3个月后,两组AFP、铁蛋白及GGT 水平均低于治疗前,且观察组低于对照组,差异有统计学意义(P <0.05)。

两组用药期间腹痛、骨髓抑制、恶心呕吐、白细胞下降等毒副反应总发生率比较,差异无统计学意义(P >0.05)。

结论:替雷利珠单抗联合贝伐珠单抗用于不可切除晚期肝癌患者中,能获得较高的近期疗效,改善AFP、铁蛋白及GGT 水平,未增加毒副反应发生率。

【关键词】 替雷利珠单抗 贝伐珠单抗 不可切除晚期肝癌 毒副反应 甲胎蛋白 doi:10.14033/ki.cfmr.2023.29.011 文献标识码 B 文章编号 1674-6805(2023)29-0041-04 Clinical Study of Tirellizumab Combined with Bevacizumab in the Treatment of Unresectable Advanced Liver Cancer/YE Tianyun, ZHANG Qihong, XU Yunpeng. //Chinese and Foreign Medical Research, 2023, 21(29): 41-45 [Abstract] Objective: To investigate the clinical effect of Tirellizumab combined with Bevacizumab in patients with unresectable advanced liver cancer. Method: A total of 96 patients with unresectable advanced liver cancer admitted to the Department of Oncology of Yangjiang People's Hospital from July 2021 to January 2023 were selected as the study objects, and they were divided into two groups by random number table method, with 48 cases in each group. The control group was treated with oral Sorafenib, and the observation group was treated with intravenous infusion of Tirellizumab combined with Bevacizumab. The short-term efficacy, biochemical indexes [alpha-fetoprotein (AFP), ferritin, gamma-glutamyl transferase (GGT)] and incidence of toxic and side effects were compared between the two groups. Result: After 3 months of treatment, the total effective rate of the observation group was higher than that of the control group, and the difference was statistically significant (P <0.05). Before treatment, there were no significant differences in biochemical indexes between the two groups (P >0.05). After 3 months of treatment, the levels of AFP, ferritin and GGT in the two groups were lower than those before treatment, and the observation group were lower than those in the control group, the differences were statistically significant (P <0.05). There was no significant difference in the total incidence of toxic and side effects such as abdominal pain, myelosuppression, nausea and vomiting, and white blood cell decline between the two groups (P >0.05). Conclusion: Tirellizumab combined with Bevacizumab in patients with unresectable advanced liver cancer can achieve high short-term efficacy, improve the levels of AFP, ferritin and GGT, and do not increase the incidence of toxic and side effects. [Key words] Tirellizumab Bevacizumab Unresectable advanced liver cancer Toxic and side effects Alpha-fetoprotein First-author's address: Yangjiang People's Hospital, Yangjiang 529500, China immune-inflammation index for overall survival of hepatocellular carcinoma patients treated with palliative Treatments[J]. J Cancer,2019,10(10):2299-2311.[13]张嘉诚,杜鹏,蒋富强,等. DC Bead 载药微球在原发性肝细胞癌患者介入治疗中的应用价值[J].中西医结合肝病杂志,2022,32(8):689-693.[14] YANG X,LI H,LIU J,et al. The short-term efficacy of DEB-TACE loaded with epirubicin and raltitrexed in the treatment of intermediate and advanced primary hepatocellular carcinoma[J]. Am J Transl Res,2021,13(8):9562-9569.[15]雒夏,刘帅伟,海龙,等.传统TACE 与DEB-TACE 治疗不可切除肝细胞癌患者的短期疗效和安全性比较[J].中华肝胆外科杂志,2023,29(3):165-169.(收稿日期:2023-08-21) (本文编辑:郎序莹) 肝癌是我国第4位常见的恶性肿瘤,其死亡率居肿瘤第2位,严重威胁居民生命与健康[1]。

动物体内转染(Entranster)答疑

动物体内转染(Entranster)答疑

动物体内转染答疑----用RNA或DNA直接注射动物完成干扰和表达动物体内转染,简单地说,就是用RNA和DNA直接打动物完成干扰和表达。

再通俗地说,用合成的(RNA)或者提取的核酸(DNA),就可以完成以前的动物转基因或者基因敲除的实验,无需再用病毒或者基因敲除动物。

实验周期可以缩短为几天,花费几千元即可进行实验。

动物体内转染技术的出现,让广大生物医学研究者,轻松进行动物的基因干扰、导入等操作。

尤其是临床医学工作者,可以在很少工作量较少经费的情况下,直接针对研究的疾病进行动物实验,发表高水平文章。

比如在英格恩客户已发表的文章中,有尾静脉注射DNA研究治疗病毒性心肌炎,有皮下肿瘤注射miRNA 研究治疗结肠癌,有脑室注射siRNA研究脑缺血机理,有皮肤涂抹siRNA治疗皮肤瘢痕等。

这些研究都非常有临床和现实意义。

由于动物体内转染技术应用的时间不长,对这种崭新的技术人们还不太了解。

此次受丁香园邀请,特开此动物体内转染相关实验技术答疑专帖。

对站友们提出的问题给予解答,希望能够和大家相互学习,共同进步。

任何与动物体内转染有关的问题(包括实验设计、产品、实验过程、结果分析、文献等问题),大家尽管提出,我们会尽力解答,也欢迎站友们参加讨论。

技术资料目录:1.体内转染试剂的原理和方法1).动物体内转染技术可以做什么?2).体内转染的原理3).体内转染的过程4).体内转染需要的实验条件5).体内转染适合进行怎样的实验6).体内转染可以在哪些组织器官进行7).应用体内转染试剂发表的部分文献8).动物体内转染和病毒感染的比较9).动物体内转染和基因敲除的比较2.体内转染实验的设计1).需要的材料2).需要的时间3).需要的费用4).常见的结果检测方法3.体内转染过程相关问题及解答1).体内转染试剂对动物有什么影响?2).注射后,试剂是如何在体内分布的,有靶向性吗?3).转染试剂和核酸需要使用多少提问与解答:1、如何技术上解决(排除)RNAi的非特异性是否需要复原实验(Rescue Experiment)。

载药脂质体包封率测定方法的研究进展_NormalPdf

载药脂质体包封率测定方法的研究进展_NormalPdf

Journal of China Pharmaceutical University2021,52(2):245-252学报载药脂质体包封率测定方法的研究进展张艺,杭太俊,宋敏*(中国药科大学药物分析系,南京210009)摘要载药脂质体已经成为改善药物体内行为或实现靶向给药的重要新剂型,对其关键工艺过程和相应的关键质量属性进行研究和控制是载药脂质体研究的重要工作。

包封程度的高低直接决定药物在体内的疗效,因此,包封率是脂质体的关键质量属性之一。

本文对常用的包封率测定方法及其特点进行分析与比较,探讨了包封率测定时需考虑的主要因素,为载药脂质体剂型开发提供参考。

关键词脂质体包封率;离心法;超滤离心法;葡聚糖凝胶柱法;微柱离心法;透析与反透析法中图分类号R944;R917文献标志码A文章编号1000-5048(2021)02-0245-08doi:10.11665/j.issn.1000-5048.20210214引用本文张艺,杭太俊,宋敏.载药脂质体包封率测定方法的研究进展[J].中国药科大学学报,2021,52(2):245–252.Cite this article as:ZHANG Yi,HANG Taijun,SONG Min.Progress in research on the determination of entrapment efficiency of liposomes [J].J China Pharm Univ,2021,52(2):245–252.Progress in research on the determination of entrapment efficiency of lipo⁃somesZHANG Yi,HANG Taijun,SONG Min*Department of Pharmaceutical Analysis,China Pharmaceutical University,Nanjing210009,ChinaAbstract Liposome,a new dosage form,has become important in improving in vivo behavior of drugs or realiz⁃ing targeted drug delivery.Study and control of its critical processes and quality attributes are the main challenges in the current research on liposomes.The degree of encapsulation can determine drug's effect in vivo directly, thus entrapment efficiency(EE)has turned into one of the critical quality attributes of liposome.In this paper some methods commonly used for the determination of EE and their characteristics are summarized and analyzed, and the main factors to be considered for the determination are discussed.Key words liposome entrapment efficiency;centrifugation;centrifugation ultrafiltration;Sephadex gel column;microcolumn centrifugation;dialysis and reverse dialysis脂质体是指将药物包封于脂质双分子层中形成的具有纳米结构的新型制剂。

体外(exvivo)体内(invivo)噬菌体展示技术—有望助力研发新一代肿瘤治疗抗...

体外(exvivo)体内(invivo)噬菌体展示技术—有望助力研发新一代肿瘤治疗抗...

体外(exvivo)体内(invivo)噬菌体展⽰技术—有望助⼒研发新⼀代肿瘤治疗抗...基于单克隆抗体(mAb)的肿瘤免疫治疗,尤其是针对免疫细胞检验点的调节治疗,在近些年来被认为是最具潜⼒和最受瞩⽬的肿瘤治疗策略之⼀。

据统计,截⾄2018年,FDA共批准31款肿瘤治疗单抗药物,其中7款单抗药物⽤于治疗⾎液性肿瘤,24款单抗药物⽤于治疗实体瘤。

在24款治疗实体瘤的单抗药物中,有9款单抗靶向了6种不同的CD抗原(包括CD19,CD20,CD30,CD33,CD38和CD53),13款单抗靶向肿瘤细胞表⾯分⼦(HER2, EGFR, PD-L1, GD2, PMSA, 以及 SLAMF7),还有3款单抗靶向免疫细胞表⾯抑制性受体PD-1和CTLA-4,⽤于激活免疫反应,剩余的单抗主要⽤于抑制肿瘤⾎管⽣成1)。

这些数据表明,⽬前可以⽤于单抗治疗药物研发的靶点是⾮常有限的,并且其中的⼤部分靶点也同时表达在正常的组织中,这意味着在单抗治疗中将会引⼊⼀定的副作⽤。

因此,如何找到新的筛选⽅法⽤于发现新的肿瘤特异性抗原,尤其是在肿瘤组织中表达量低,但却发挥重要作⽤的抗原,以及靶向这些抗原的抗体,对于扩⼤单抗在肿瘤治疗领域中的应⽤具有重要的意义。

本⽂将要介绍的体外 (ex vivo) / 体内 (in vivo) 噬菌体展⽰技术,主要⽬的就是为了找到更多的肿瘤特异性抗原及靶向抗原的抗体。

这些抗原应该具有的特点包括:低丰度;表达在罕见的亚细胞群表⾯;特异地存在于肿瘤微环境中。

因此有望为开发新⼀代肿瘤治疗抗体提供帮助。

那么这些新的筛选体系是怎么做到的呢?我们知道,鉴定肿瘤抗原的传统⽅法是在DNA, RNA 和蛋⽩质⽔平上寻找肿瘤和⾮肿瘤细胞的区别2)。

这依赖于有效⽣物原材料的获取,⽐如细胞系和临床样品。

然⽽,抗原在细胞系表⾯的表达⽔平并不能代表其在肿瘤组织中的表达⽔平。

有研究表明,⼀些细胞表⾯表达的分⼦在从体内分离到体外的过程中会失去其上调表达的特征,从⽽会导致⼀些潜在靶点的丢失3)。

纳米药物递送系统应用于肿瘤免疫治疗的研究进展

纳米药物递送系统应用于肿瘤免疫治疗的研究进展

广东药科大学学报Journal of Guangdong Pharmaceutical University Jul,2023,39(4)收稿日期:2023-04-03基金项目:河北省自然科学基金面上项目(C2019203556)作者简介:杨逸博(1999-),男,硕士研究生,主要从事纳米药物递送系统在化疗与免疫联合治疗中的研究,Email :*****************通信作者:李健(1976-),博士,副教授,主要从事非编码RNA 与肿瘤发生相关机制研究、抗肿瘤药物靶向性转运载体的构建、基于核酸适配体的肿瘤早期诊断试剂盒的研究与应用,Email :*****************.cn 。

纳米药物递送系统应用于肿瘤免疫治疗的研究进展杨逸博,李健(燕山大学环境与化学工程学院,河北秦皇岛066000)摘要:癌症免疫治疗是一种倍受关注的治疗策略。

然而,免疫治疗面临的主要挑战包括患者反应性低、肿瘤特异性差、存在免疫抑制性肿瘤微环境等。

纳米药物递送系统(nano drug delivery systems,NDDS )被用于负载药物,经修饰后可表现出肿瘤靶向性给药、肿瘤微环境响应和位点特异性释放等优异性能。

因此,NDDS 可以被有效地用于癌症免疫治疗,能减少毒副作用和免疫相关抑制。

本文重点介绍了近来基于NDDS 的免疫治疗的研究进展,包括诱导免疫原性细胞死亡(immunogenic cell death,ICD )、联合肿瘤免疫检查点抑制剂促进免疫治疗疗效、改善肿瘤免疫抑制微环境3个方面。

关键词:纳米药物递送系统;肿瘤细胞;免疫原性细胞死亡;免疫检查点;肿瘤微环境中图分类号:R94文献标识码:A文章编号:2096-3653(2023)04-0135-08DOI :10.16809/ki.2096-3653.2023040302Research progress of nano drug delivery systems in tumor immunotherapyYANG Yibo,LI Jian *(College of Environmental and Chemical Engineering,Yanshan University,Qinhuangdao 066000,China )*Corresponding author Email:*****************.cnAbstract:Cancer immunotherapy is an attractive therapeutic strategy.However,the main challenges faced by immunotherapy include low patient responsiveness,poor tumor specificity,existence of immunosuppressive tumor microenvironment,etc.Nano drug delivery systems (NDDS)have been applied to load drugs extensively.After modification,NDDS exhibit excellent performances,such as tumor targeted drugs,tumor microenvironment response and site-specific release.Therefore,NDDS can be effectively used in cancer immunotherapy to reduce toxic side effects and immune related suppression.In this review,we focused on the recent research progress of immunotherapy based on NDDS,including the induction of immunogenic cell death (ICD),the combination of tumor immune-checkpoint inhibitors to promote the efficacy of immunotherapy,and the improvement of tumor immune suppression microenvironment.Key words:nano drug delivery system;tumor cell;immunogenic cell death;immune checkpoint block;tumor microenvironment目前癌症仍是全球病患死亡的主要原因,且发病率逐年上升[1,2],癌症治疗研究备受关注。

siRNA的体内递送

siRNA的体内递送

siRNA的体内递送师明磊,赵志虎*,王洋,陈惠鹏军事医学科学院生物工程研究所,北京 100071摘要:RNA干扰是一种由小干扰RNA介导的转录后基因沉默。

自利用RNAi沉默目的基因获得成功以来,体内应用RNAi 的研究受到高度重视。

由于siRNA本身的不稳定性以及体内的复杂环境,siRNA递送的安全性与有效性成为目前关注的重点。

文章就目前报道的siRNA体内的递送方式进行了综述。

关键词:RNAi; siRNA; siRNA递送In vivo delivery of siRNASHI Ming-Lei, ZHAO Zhi-Hu, WANG Yang, CHEN Hui-PengBeijing Institute of Biotechnology, Beijing 100071, ChinaAbstract: RNA interference (RNAi) is a mechanism of posttranscriptional gene silencing mediated by small interfering RNA (siRNA). The ability of synthetic siRNA to silence genes in vivo has made it well suited as therapeutic drug, but the instability and polarity of siRNA and the complexity of in vivo circumstances retarded rapid development of RNAi-based therapies. In this review, a summary of the advances in in vivo siRNA delivery is presented and discussed.Keywords: RNAi; siRNA; siRNA deliveryRNA干扰(RNA interference, RNAi),是一种能够有效抑制基因表达的靶向性转录后基因沉默机制[1]。

治疗癌症的奇思妙想的作文

治疗癌症的奇思妙想的作文

治疗癌症的奇思妙想的作文英文回答:Cancer, a word that strikes fear into the hearts of many. It is a disease that has claimed the lives of countless individuals and their loved ones. Finding a cure for cancer has been a goal for scientists and researchers for decades. While there have been significant advancements in cancer treatment, there is still much more to be done.One unconventional idea that I have for treating cancer is using nanotechnology. Nanotechnology involves the manipulation of materials at the nanoscale, which is about 1 to 100 nanometers in size. By utilizing nanotechnology, we can potentially target cancer cells directly and deliver drugs or therapies with greater precision.For example, imagine tiny nanoparticles that are designed to specifically seek out and destroy cancer cells. These nanoparticles could be injected into the body andguided to the tumor site using magnetic fields or other techniques. Once at the tumor site, the nanoparticles could release a drug or therapy that specifically targets the cancer cells, leaving healthy cells unharmed.Another idea is to harness the power of the immune system to fight cancer. Immunotherapy has shown promising results in recent years, with the development of drugs that can stimulate the immune system to recognize and attack cancer cells. One such example is the use of checkpoint inhibitors, which block proteins that prevent immune cells from attacking cancer cells. By removing these checkpoints, the immune system can effectively target and destroy the cancer cells.In addition to these ideas, there is also the potential for personalized medicine in cancer treatment. Each individual's cancer is unique, and what works for one person may not work for another. By analyzing the genetic makeup of a patient's tumor, we can tailor treatment options to their specific needs. This could involve using targeted therapies that are designed to attack specificgenetic mutations or using a combination of treatments that have been shown to be effective for similar cases.中文回答:癌症,这个词让许多人心生恐惧。

非小细胞肺癌中microRNAs与EGFR-TKIs继发性耐药机制的研究进展

非小细胞肺癌中microRNAs与EGFR-TKIs继发性耐药机制的研究进展

中国肺癌杂志2014年12月第17卷第12期Chin J Lung Cancer, December 2014, Vol.17, No.12·860··综述·非小细胞肺癌中microRNAs 与EGFR-TKIs继发性耐药机制的研究进展段晓阳 史健【摘要】 近年来,在非小细胞肺癌(non-small cell lung cancer, NSCLC )靶向治疗中,尤其是伴有表皮生长因子受体(epidermal growth factor receptor, EGFR )基因突变的患者,EGFR 酪氨酸激酶抑制剂(tyrosine kinase inhibitor, TKI )越来越多地进入到临床治疗,但EGFR-TKI 耐药的产生不仅影响药物敏感性,甚至出现疾病进展,成为制约其疗效的主要瓶颈。

微小RNA (microRNAs, miRNAs )是一种非编码蛋白的RNA ,参与转录后水平基因的表达调控,最近研究发现,miRNAs 参与了EGFR-TKIs 耐药,影响肿瘤细胞对吉非替尼的敏感性。

本文就NSCLC 中miRNAs 与EGFR-TKIs 继发性耐药之间的相关性研究进展做简要的综述。

【关键词】 肺肿瘤;MicroRNAs ;EGFR-TKIs ;继发性耐药Advance in MicroRNAs and EGFR-TKIs Secondary Resistance Researchin Non-small Cell Lung CancerXiaoyang DUAN 1, Jian SHI21Graduate Student of Hebei Medical University, Shijiazhuang 050000, China; 2Department of Medical Oncology,Hebei Province Cancer Hospital, Shijiazhuang 050000, ChinaCorresponding author: Jian SHI, E-mail: shijian6668@【Abstract 】 In recent years, in non-small cell lung cancer (NSCLC) targeted therapy, especially in patients with epidermal growth factor receptor (EGFR ) mutations, EGFR-tyrosine kinase inhibitors (TKI) more and more come into the clinical treatment, but EGFR-TKI resistance not only influence the drug sensitivity, appear even disease progression, become the main bottleneck of its curative effect. MicroRNAs (miRNAs) is a non coding RNA and protein involved in regulating gene expression in the transcription level. Recent studies found that miRNAs involved in EGFR-TKIs resistance, which affect the sensitivity of tumor cells to treatment. In this paper, we reviewed briefly advance in miRNAs and EGFR-TKIs secondary resis-tance research in NSCLC.【Key words 】 Lung neoplasms; MicroRNAs; EGFR-TKIs; Secondary resistanceDOI: 10.3779/j.issn.1009-3419.2014.12.07作者单位:050000 石家庄,河北医科大学(段晓阳);050000 石家庄,河北医科大学第四医院肿瘤内科(史健) (通讯作者:史健,E-mail: shijian6668@ )近几年,肺癌在发达及发展中国家的发病率逐年增高,即使在发达国家,5年相对生存率也仅为16%[1]。

用于肿瘤靶向性X线CT造影剂和抗肿瘤药物传递的多功能含碘纳米粒子的制备和表征

用于肿瘤靶向性X线CT造影剂和抗肿瘤药物传递的多功能含碘纳米粒子的制备和表征

用于肿瘤靶向性X线CT造影剂和抗肿瘤药物传递的多功能含碘纳米粒子的制备和表征朱颖;张岩;陈研;杨晓英【摘要】目的:制备用于肿瘤靶向性X线CT造影剂和抗肿瘤药物传递的多功能含碘纳米粒子,用于肿瘤诊断和治疗.方法:利用沉淀聚合法制备含碘聚合物纳米粒子P(MATIB-co-MBA-co-GMA)-FA-AuNP,该纳米粒子以2-甲基丙烯酰(3-酰胺-2,4,6-三碘苯甲酸)(MATIB)为单体,以N,N’-亚甲基双丙烯酰胺为交联剂(MBA),通过甲基丙烯酸缩水甘油酯(GMA)和乙二胺(EDA)将叶酸分子修饰到该纳米粒子表面,并原位沉积金纳米粒子(AuNP).结果:TEM结果显示该纳米粒子分散均匀,平均粒径为135 nm.体外X线CT成像检测结果表明AuNP的掺杂显著增加了该纳米粒子的X线衰减性能.该纳米粒子同时可高效负载抗肿瘤药物(DOX),载药量为51.3%,并具有pH敏感的可控释放性能.体外药物输送结果显示有FA修饰的纳米粒子能更好地携载抗肿瘤药物进入肿瘤细胞.细胞毒性的结果显示该P(MATIB-co-MBA-co-GMA)-FA-AuNP纳米粒子在浓度低于100 μg/mL时未显示明显毒性.载药后,有叶酸修饰的纳米粒子对肿瘤细胞具有更好的杀伤性能.结论:该纳米粒子可同时作为肿瘤靶向性X线CT造影剂和抗肿瘤药物载体,用于肿瘤诊断和治疗.【期刊名称】《天津医科大学学报》【年(卷),期】2018(024)002【总页数】9页(P122-130)【关键词】沉淀聚合;肿瘤靶向;CT造影剂;药物输送【作者】朱颖;张岩;陈研;杨晓英【作者单位】天津医科大学药学院,天津市临床药物关键技术重点实验室,天津300070;天津医科大学第二医院药学部,天津300211;天津医科大学药学院,天津市临床药物关键技术重点实验室,天津300070;天津医科大学药学院,天津市临床药物关键技术重点实验室,天津300070;天津医科大学药学院,天津市临床药物关键技术重点实验室,天津300070【正文语种】中文【中图分类】O63计算机断层扫描(CT)是目前临床上应用最广泛的非侵入性成像诊断技术之一,在临床已经使用大半个世纪。

IVIVE(invitrotoinvivoextrapolation体外-体内外推法)

IVIVE(invitrotoinvivoextrapolation体外-体内外推法)

IVIVE(invitrotoinvivoextrapolation体外-体内外推法)在药物临床开发前,准确的预测人体的肝清除率十分重要且较难。

IVIVE是一种较常见的方法,即根据肝微粒体或肝细胞测得体外固有清除率CLint,再引入MPPGLHPGLliverweightbodyweighthepaticbloodflow等calingfactor,通常采用well-tirred模型计算得到体见下文。

目前IVIVE法突出的问题包括两个:1、采用肝微粒体还是肝细胞的代谢稳定性数据进行计算;2、经常出现低估现象,如何校正外推值使其更接近实测值。

肝微粒体易制备、保存的特点,为IVIVE的最常用方法。

理论上而言,肝细胞相比较于肝微粒体更适合IVIVE,因为肝细胞含有肝脏转运体和更多的代谢酶,更接近体内生理条件。

但研究表明,在转运体无显著的主动摄取的前提下,对于CYP3A4的底物而言,肝微粒体的预测值比肝细胞更准确。

对于CYP3A4的底物,肝微粒体的CLint测定值常高于肝细胞;而对于CYP2C的底物两者无明显差别;对于UGT的底物,低清除率条件下,两者接近,随着清除率增加,肝细胞值较高。

对于该现象,猜测可能是由于tranporter-enzyme的相互作用仅存在于肝细胞中,CYP3A4和P-gp有着相似的底物特异性,认为药物在进入肝细胞或者肠上皮细胞将要被代谢时,P-gp将其外排避免代谢,由此导致CYP3A4的底物肝细胞CLint偏低。

Lamandbenet发现elacridar(P-gp抑制剂)在微粒体中对地高辛的代谢无影响,但是在肝细胞中增加了其代谢。

此外,CLint的大小与预测的准确性也有一定的关联。

高清除率的化合物,当其为CYP2D6的底物时,microome与hepatocyted的差别不显著;为CYP3A4的底物时差异被放大。

小结:1、CYP3A4的底物肝微粒体的Clint的测定值通常高于肝细胞,无明显主动摄取转运体作用时,肝微粒数据进行IVIVE准确性更高;2、对于CYP3A4的底物,高清除率的化合物微粒体与肝细胞的差异明显;低清除率的化合物差异较小;3、即使CYP3A4底物采用肝微粒体预测的误差相比于肝细胞较低(5.19forhepatocyteand3.47formicroome),但仍然处于较高的水平,个人常在推算后取3作为校正因子;4、对于CYP1A2的底物发现采用肝微粒体和肝细胞预测的准确性均较高。

mir-21的转录调控及在肿瘤中的作用研究进展

mir-21的转录调控及在肿瘤中的作用研究进展

mir-21的转录调控及在肿瘤中的作用研究进展张经波【期刊名称】《重庆医学》【年(卷),期】2012(041)031【总页数】3页(P3336-3337,3346)【关键词】mir-21;微RNAs;肿瘤【作者】张经波【作者单位】第三军医大学西南医院消化内科,重庆,400038【正文语种】中文微RNAs(miRNAs)是一类在转录后水平调控基因表达的小的内源性非编码RNA 分子,约长17~27 nt,在细胞与组织中特异表达。

miRNAs在各种不同的生物学进程中都发挥关键作用,通过靶向结合基因中与成熟miRNA互补的序列,调控发育、细胞分化、增殖和凋亡。

miRNAs-靶基因间调控的异常与一系列癌症的进程密切相关。

超过50%的人类miRNAs基因位于脆性位点或与癌症相关的区域,意味着miRNAs在癌症的发生发展中可能扮演重要角色。

一些miRNAs在癌细胞与组织中异常表达,并与肿瘤的类型及进程相关。

miRNAs的靶基因通常为癌基因或肿瘤抑制基因,通过异常调控miRNAs-靶基因间关系及miRNAs自身表达可直接或间接影响着癌症的命运[1]。

mir-21符合了上面所叙述miRNA的所有特点,并作为较早研究的miRNAs之一得到越来越多的关注。

本文就已经研究比较详尽的mir-21有关基因定位、生成、表达调控及与肿瘤的关系作一综述。

1 mir-21的基因定位mir-21在包括哺乳动物、鸟类和鱼类在内的一系列脊椎动物中都表现出较强的进化保守性。

人类中mir-21的基因定位首次被发现定位于17q23.2,位于TMEM49基因的第10个内含子内。

虽然转录mir-21的启动子位点与蛋白编码基因TMEM49内含子有重叠序列,但是pri-mir-21(mir-21的初始转录体)却是被一个保守的启动子独立转录的[2]。

2 mir-21的转录成熟的mir-21来源于3 433 nt长的pri-mir-21,在pri-mir-21转录体的+3 394与+3 399间有一个保守的AAU AAA多聚腺苷酸化信号,pre-mir-21(mir-21的颈环前体)则位于+2 445与+2 516间。

miRNA(医学免疫学)

miRNA(医学免疫学)

Kim and Nam 2006
miRNA profile in colon cancer stem cells VS non-stem cells
Fang and Zhou 2012 Oncology Reports
Today’s topics
What is miRNA? How are miRNAs discovered? How are miRNAs synthesized in vivo? How many miRNAs are there? Where are miRNAs located in the genome? When and where are miRNAs expressed? How miRNAs regulate gene expression? What genes do miRNAs regulate? What regulate miRNAs? What are the general functions of miRNAs? Is there any therapeutic application?
Mechanism of function
Up-regulation VS down regulation Transcription level VS post-transcription level Cleavage VS translational repression Translational repression at the initiation step Translational repression post-initiation
Discovery of miRNA
“Attempted to overexpress chalcone synthase (CHS) in pigmented petunia petals by introducing a chimeric petunia CHS gene. Unexpectedly, the introduced gene created a block in anthocyanin biosynthesis.”

HASTY moves to chromatin for miRNA production

HASTY moves to chromatin for miRNA production

CelPress Partner Journal Molecular PlantSpotlightHASTY moves to chromatin for miRNA productionMicroRNAs (miRNAs) have been well appreciated for their critical roles in regulating various biological processes in eukaryotes since the initial discovery of these tiny molecules in 1990s. The factory for miRNAs is located inside the nucleus, whereas their workplace is the cytoplasm. Thus, the prompt transport of miR­NAs from nucleus to cytoplasm is essential for their proper func­tionality. In metazoans, the commitment is carried out by Exportin5 (EXP5/XP05), the nuclear exporter of miRNA precursors (pre- miRNAs) (Yi et al., 2003). In plants, HASTY (HST), a karyopherin family member that shuttles between nucleus and cytoplasm, has been identified as a component in the miRNA pathway (Park, et al., 2005). Since HST is an ortholog of mammalian EXP5/XP05, the protein has been ambiguously assumed to act as an exporter of plant miRNAs. However, a controversial observation is that the subcellular distribution of miRNAs between nucleus and cytoplasm is not altered, despite the decrease in their overall steady-state levels in the hst mutants (Park et al., 2005). Furthermore, it has been shown that AG01, a master effector in plant RNA silencing, moves into nucleus to uptake miRNAs and subsequently returns to cytoplasm for functions via EXPORTIN1 (EXP01/CRM1) (Bologna et al., 2018), and that the trafficking of AG01-miRNA complexes is promoted by Nucleoporin 1(NUP1)/Transcription-coupled Export 2 com­plex (TREX-2) (Zhang et al., 2020). These reports also challenged HST function as a miRNA exporter. An excellent study by Cambiagno et al. (2020), recently published in Molecular Plant, solved the long-term mystery and demonstrated that HST orchestrates the transcription and processing of primary substrates of miRNAs (pri-miRNAs), rather than serving as an exporter for miRNAs as previously thought (Figure 1).Cambiagno and colleagues began to address the issue by asking whether the exporting function of HST is essential for its role in the miRNA pathway. Karyopherins typically conduct cellular transport in cooperation with the small GTPases RANs. Similarly, HST, through its N-terminal domain, interacts with the GTPases RAN 1-3 for the potential exporting activity. Cambiagno and colleagues did not observe any change in the steady-state accumulation or nuclear/cytoplasmic ratios of tested miRNAs in ran single or double mutants relative to Col- 0. Similar to ran mutants, complementation transformants ex­pressing N-terminal domain depleted HST (hst/HSTA N) did not show a change in subcellular distribution of miRNAs. However, hst; HST jn harbored, similar to hst itself, reduced levels of miRNAs. Hence, the initial results led the authors to conclude that HST positively regulates miRNA accumulation via its N-terminal domain and this function can be uncoupled from HST exporting activity. Next, Cambiagno and colleagues as­sessed subcellular localizations of HST truncation forms in different backgrounds. Full length HST displayed an increased nucleus/cytoplasm ratio in ran7, consistent with the proposed role of RAN1 in the HST shuttling. Surprisingly, HSTAN,which does not interact with RAN proteins, displayed a decreased nucleus/cytoplasm distribution in Col-0. The contrasted cellular364 Molecular Plant 14, 364-365, March 12021 © The Author 2021.localizations of HST and HSTA N implied the presence of an im- portin that moves HST to the nucleus through interaction with the N-terminal domain of HST. Cambiagno and colleagues iden­tified the importin protein as an a-importin I MPA-2 through mass spectrometry. Importantly, the authors found the steady- state levels of miRNAs, but not their distributions in nucleus/ cytoplasm, were reduced in impa-2relative to Col-0. These re­sults prompted the authors to predict a possible regulatory role of nuclear HST in miRNA biogenesis.Cambiagno and colleagues then performed high-throughput sequencing of small RNAs and pri-miRNAs. Notably, the downre- gulated miRNAs observed in hst showed a higher overlapping with those in microprocessor mutants like hyl1and se, but not the ones in downstream mutants. Cambiagno and colleagues also detected the over-accumulation of pri-miRNAs in the hst mutant. Furthermore, the pri-miRNA profile in hst showed the strongest correlation with that of dcl1-9compared with other miRNA mutants. Despite this, HST did not affect DCL1 activity via a semi-/'n vivo microprocessor assay. Thus, HST affects pri- miRNA accumulation in a way different from mammalian EXP5/ XP05 that could also facilitate Drosha/DGCR8 processing activ­ity independent of its exportin function (Wang et al., 2020). A ribonucleoprotein immunoprecipitation (RIP) assay showed that HST, but not HSTAN,could associate with mature miRNAs. These observations led the authors to assess possible factors guiding the interaction between HST and miRNAs. Cambiagno and colleagues pinpointed DCL1,but not other components of the microprocessor, as a HST partner through bimolecular fluorescence complementation (BiFC) and yeast-two-hybrid screenings. Moreover, the interaction with DCL1 is through the N-terminal domain of HST.Transcription and processing of the pri-miRNAs are coupled pro­cesses as many factors involved in miRNA biogenesis are re­cruited to MIR loci (Wang et al., 2018). Also, MEDIATOR complex (MED) is essential for pri-miRNA transcription by recruit­ing RNA Pol II to MIR gene promoters (Kim et al., 2011). Indeed, Cambiagno and colleagues recovered four subunits of MED37- C/D/E/F among several RNA binding proteins in their mass spec­trometry datasets of HST. The authors then hypothesized that HST, MED37, and DCL1 could form a complex at MIR gene loci to orchestrate pri-miRNA transcription and processing. The inter­actions among the proteins were validated through a series of BiFC, triple-molecular fluorescence complementation and yeast-three-hybrid assays. Furthermore, Cambiagno and col­leagues detected the binding of MED37 complex to several MIR gene loci. Importantly, the authors found that the interaction between DCL1 and MIR loci could only be present in Col-0 but not in hst.Altogether, the results indicated that HST promotesPublished by the Molecular Plant Shanghai Editorial Office in association with Cell Press, an imprint of Elsevier Inc., on behalf of CSPB and CEMPS, CAS.Spotlight Molecular PlantFigure 1. HST m oves c lo se to M IR lo ci fo rm iR N A p ro d u c tio n.Transcription of MIR loci is positively regulated byMED complexes. Pri-miRNAs are processed byDCL1 -centered microprocessors to producemiRNAs, which are in turn exported from the nu­cleus through AG01-RISC complex. Cambiagnoand colleagues found that, through the interactionwith MED complexes and DCL1, HST recruitsDCL1 to MIR loci and facilitates the capture andprocessing of pri-miRNAs. HST is imported fromcytoplasm to nucleus by IMPA-2 and exported tocytoplasm by RAN1 without mature miRNAs. Thebona fide cargos of HST remain to be identified.MIR, MIR genes; MED, Mediator complex; DCL1,Dicer-Like 1; SE, Serrate; HYL1, HyponasticLeaves 1; IMPA-2, Importin a Isoform 2; RAN1,Ras-Related Nuclear Protein 1; A G01, Argonaute1; EXP01, Exportin 1.the association of DCL1 to M IR genes and stabilizes the DCL1 - MED37 complex at M IR loci.The work by Cambiagno et al. (2020) stands out because it clarifies the early confusion of HST function as an exportin of miRNAs. Moreover, their study provides compelling evidence that HST serves as a platform regulating M IR transcription and facilitating pri-miRNA processing. This work not only expands the potential functions of the karyopherin family in plants, but also raises many interesting questions for follow-up investigation. First, since HST shares all features of exportin proteins, what are the bona fide car­gos for HST? Given HST interacts with DCL1 and MED complexes, it might regulate nucleocytoplasmic trafficking of these compo­nents, which in turn affects miRNA biogenesis. If so, this would be reminiscent of an importin, KETCH 1, which controls the HYL1 distribution between nucleus and cytoplasm (Zhang et al., 2017). Second, is there trafficking of miRNAs/miRNA*s (complementary strands of miRNAs) and small interfering RNAs (siRNAs) and their passenger strands across the nuclear pores in plants? Small RNAs, once produced, are typically housed and protected in RNA-inducing silencing complex (RISC). A rabidopsis does harbor AG01-free but residual miRNAs (Wang et al., 2011). Given this circumstance, do HST or other karyopherin members contribute to the trafficking of the A G01 -free miRNAs, also as the authors sus­pected, or even siRNAs? Third, are there any additional karyo­pherin members that contribute to miRNA functions or biogenesis? On the other hand, since the recruitment of HST to M IR loci is dependent on MED complexes, does HST regulate the transcrip­tion of loci such as protein-coding genes and long noncoding RNAs beyond M IR genes? Research on these topics will surely flourish in the future.ACKNOWLEDGMENTSThe work by the Zhang laboratory was supported by a grant from NIH (GM132401) to X.Z. No conflict of interest declared.Di Sun and Xiuren Zhang* Department of Biochemistry and Biophysics. Texas A&M University, CollegeStation, TX 77843, USA^Correspondence:XiurenZhang(*********************)https:///10.1016/j.molp.2021.01.012 REFERENCESBologna, N.G., Iselin, R.f Abriata, L.A., Sarazin, A., Pum plin, N., Jay, F., Grentzinger, T., Dal Peraro, M., and Voinnet, O. (2018). Nucleo- cytosolic shuttling of ARGONAUTE1 prompts a revised model of the plant microRNA pathway. Mol. Cell 69:709-719.e5.Cambiagno, D.A., G iudicatti, A.J., Arce, A.L., Gagliardi, D., Li, L., Yuan, W., Lundberg, D.S., Weigel, D., and Manavella, P.A. (2020).HASTY modulates miRNA biogenesis by linking pri-miRNA transcription and processing. Mol. Plant https:///10.1016/j.molp.2020.12.019.Kim, Y.J., Zheng, B., Yu, Y., Won, S.Y., Mo, B., and Chen, X. (2011). The role of Mediator in small and long noncoding RNA production in Arabidopsis thaliana.EMBO J. 30:814-822.Park, M.Y., Wu, G., Gonzalez-Sulser, A., Vaucheret, H., and Poethig, R.S. (2005). Nuclear processing and export of microRNAs in Arabidopsis.Proc. Natl. Acad. Sci. U S A 102:3691-3696.Wang, H., Zhang, X., Liu, J., Kiba, T., Woo, J., Ojo, T., Hafner, M., Tuschl, T., Chua, N.H., and Wang, X.J. (2011). Deep sequencing of small RNAs specifically associated with Arabidopsis AG01 and AG04 uncovers new AGO functions. Plant J. 67:292-304.Wang, J., Lee, J.E., Riemondy, K., Yu, Y., Marquez, S.M., Lai, E.C., and Yi, R. (2020). XP05 promotes primary miRNA processing independently of RanGTP. Nat. Commun. 11:1845.Wang, Z., Ma, Z., Castillo-Gonzalez, C., Sun, D., Li, Y., Yu, B., Li, P., and Zhang, X. (2018). SWI2/SNF2 ATPase CHR2 remodels pri- miRNAs via SE to impede miRNA production. Nature 557:516-521.Yi, R., Qin, Y., Macara, I.G., and Cullen, B.R. (2003). Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 17:3011-3016.Zhang, B., You, CM Zhang, Y., Zeng, L., Hu, J., Zhao, M., and Chen, X.(2020). Linking key steps of microRNA biogenesis by TREX-2 and the nuclear pore complex in Arabidopsis.Nat. Plants 6:957-969.Zhang, Z.H., Guo, X.W., Ge, C.X., Ma, Z.Y., Jiang, M.Q., Li, T.H., Koiwa,H., Yang, S.W., and Zhang, X.R. (2017). KETCH1 imports HYL1 tonucleus for miRNA biogenesis in Arabidopsis.Proc. Natl. Acad. Sci.U S A 114:4011-4016.Molecular Plant 14, 364-365, March 12021 © The Author 2021.365。

治疗癌症的奇思妙想的作文

治疗癌症的奇思妙想的作文

治疗癌症的奇思妙想的作文英文回答:Cancer is a devastating disease that affects millions of people worldwide. As a researcher, I have been pondering on unconventional ideas to treat this deadly condition. One of my innovative thoughts revolves around the concept of using nanotechnology to target and destroy cancer cells.Nanotechnology involves the manipulation of matter at the nanoscale, which is one billionth of a meter. By engineering nanoparticles to specifically seek out and attach to cancer cells, we can deliver targeted therapies directly to the tumor site. These nanoparticles can be loaded with drugs or other therapeutic agents that will selectively kill the cancer cells, while leaving healthy cells unharmed.The use of nanotechnology in cancer treatment has shown promising results in preclinical studies. For example,researchers have successfully used gold nanoparticles to deliver chemotherapy drugs directly to tumor cells in mice. This approach not only increased the effectiveness of the treatment, but also reduced the side effects associated with traditional chemotherapy.Furthermore, nanotechnology can also be utilized to detect cancer at its earliest stages. By developing sensitive nanosensors, we can detect the presence of cancer biomarkers in the blood or other bodily fluids. This early detection can significantly improve the chances of successful treatment, as the cancer can be diagnosed and treated before it spreads to other parts of the body.In addition to nanotechnology, another unconventional idea I have been exploring involves the use of immunotherapy for cancer treatment. Immunotherapy harnesses the power of the immune system to recognize and destroy cancer cells. This approach has shown remarkable success in certain types of cancer, such as melanoma and lung cancer.One form of immunotherapy that has gained attention ischeckpoint inhibitors. These drugs block the proteins that prevent immune cells from attacking cancer cells. By inhibiting these proteins, we can unleash the fullpotential of the immune system to fight against cancer.Another approach in immunotherapy is adoptive cell transfer, where immune cells are extracted from the patient, genetically modified to enhance their cancer-fighting abilities, and then reintroduced into the patient's body. This personalized treatment has shown promising results in clinical trials, with some patients experiencing long-term remission.In conclusion, thinking outside the box is essential when it comes to finding new ways to treat cancer. The useof nanotechnology and immunotherapy are just two examplesof unconventional approaches that hold great potential in the fight against this devastating disease.中文回答:治疗癌症是一个全球范围内影响数百万人的可怕疾病。

活细胞药物体内可视化研究进展

活细胞药物体内可视化研究进展

学报Journal of China Pharmaceutical University2022,53(2):156-163156活细胞药物体内可视化研究进展魏奶杰,王广基*,张经纬**(中国药科大学江苏省药物代谢动力学重点实验室,南京210009)摘要活细胞药物的研发以及治疗的成功实施均需要充分阐明移植后的细胞命运,这对活细胞药物的有效性和安全性至关重要。

为了解决这一问题,细胞成像技术进入人们视线,利用可视化技术对活细胞药物进行无创示踪,可以了解活细胞药物在体内的分布、归巢、活性等,有助于确定最佳的移植细胞数量、优化给药方案、提高移植效率、增强作用的靶向性、降低潜在的靶外积累风险。

本文从放射性核素成像技术、磁共振成像技术、磁颗粒成像技术、计算机断层扫描成像技术、荧光成像技术以及多模态成像技术综述了活细胞药物体内无创可视化示踪的研究进展,旨在指导应用合适的造影剂和示踪技术,获得活细胞药物在体内的生物学行为,为活细胞药物研发及其移植治疗提供更加合理的科学依据。

关键词活细胞药物;体内成像;CAR-T细胞;间充质干细胞;进展中图分类号R91文献标志码A文章编号1000-5048(2022)02-0156-08doi:10.11665/j.issn.1000-5048.20220204引用本文魏奶杰,王广基,张经纬.活细胞药物体内可视化研究进展[J].中国药科大学学报,2022,53(2):156–163.Cite this article as:WEI Naijie,WANG Guangji,ZHANG Jingwei.Advances in research on visualization of living cell drugs in vivo[J].J China Pharm Univ,2022,53(2):156–163.Advances in research on visualization of living cell drugs in vivoWEI Naijie,WANG Guangji*,ZHANG Jingwei**Key Labratory Jiangsu Provincial of Drug Metabolism and Pharmacokinetics,China Pharmaceutical University,Nanjing210009, ChinaAbstract The development of living cell drugs and their successful application in clinical treatments require full clarification of the fate of cells after transplantation,which is critical to the safety and efficacy of living cell drugs.In order to solve this problem,cell imaging technology has come into our sight,and the use of visualization technology for non-invasive tracing of living cell drugs could reveal the distribution,homing and activity of living cell drugs in the body,which helps to determine the best number of transplanted cells,optimize the administra‑tion scheme,improve the transplantation efficiency,enhance the targeting of transplanted cells,and reduce the potential off-target accumulation risk.This paper summarizes the research advances of non-invasive visual trac‑ing in vivo for living cell drugs from the perspectives of radionuclide imaging,magnetic resonance imaging, magnetic particle imaging,computed tomography imaging,fluorescence imaging and multimodal imaging.The aim is to obtain the biological behavior of living cell drugs in vivo with the application of appropriate contrast agent and tracing technology,and provide a more reasonable scientific basis for the research and development of living cell drugs and their transplantation therapy.Key words living cell drug;in vivo imaging;CAR-T cell;mesenchymal stem cells;advancesThis study was supported by the National Natural Science Foundation of China(No.82173887,82073928)收稿日期2021-12-03通信作者*Tel:************E-mail:guangjiwang@**Tel:************E-mail:zhangjw_cnnj@基金项目国家自然科学基金资助项目(No.82173887,No.82073928)第53卷第2期魏奶杰,等:活细胞药物体内可视化研究进展面对重大疾病精准治疗的临床紧迫需求,生物医药行业正在从传统的小分子化学药物时代进入精准靶向生物药/细胞治疗药物的个性化治疗时代。

RNAi治疗中的纳米递送系统

RNAi治疗中的纳米递送系统

RNAi治疗中的纳米递送系统发布时间:2023-06-02T11:09:10.049Z 来源:《医师在线》2023年1月2期作者:苏吴越1 乔雪1 洪学传2 [导读]RNAi治疗中的纳米递送系统苏吴越1 乔雪1 洪学传2(1.西藏大学医学院;西藏 拉萨850000,2.西藏大学理学院;西藏拉萨850000)摘要:RNAi是一种由外源性或内源性dsRNA介导的基因沉默现象,是目前最有效、最先进的基因治疗方法之一。

尽管RNAi药物的治疗潜力显而易见,但其制剂必须克服阻碍其发展为临床应用的各种障碍。

纳米技术领域的最新进展导致了新型递送系统的开发,该系统能够RNA递送至肿瘤。

在这篇综述中,我们对目前用于RNAi治疗的纳米载体进行了分类,并总结了不同纳米载体的特点。

关键词:R NA i治疗;RNA递送;纳米载体Nanodelivery systems in RNAi therapyWuyue S u 1,X ue Q iao1,X uechuan H ong2(1.Medical School of Tibet University, Lhasa, Tibet 850000,2. School of Science, Tibet University, Lhasa, Tibet850000)Abstract: RNAi is a gene silencing phenomenon mediated by exogenous or endogenous dsRNA. It is one of the most effective and advanced gene therapy methods. Despite the obvious therapeutic potential of RNAi drugs, their formulations must overcome various obstacles that prevent their development into clinical applications. Advances in nanotechnology have led to the development of novel delivery systems that deliver RNA to tumors. In this review, we classify nanocarriers currently used for RNAi therapy and summarize the characteristics of different nanocarriers.Key words: RNAi therapy; RNA delivery; Nanomedicine carrier1998年,Fire等人发现了RNA干扰(RNA interference, RNAi),为基于RNA寡核苷酸的新基因靶向方法的开发奠定了基础[1]。

小鼠心肌层注射方法的改进

小鼠心肌层注射方法的改进

小鼠心肌层注射方法的改进王惠;赵蓉;周学中【摘要】探索心肌有效稳定的给药方式,达到优化手术的均一性和可重复性.采用改良后的注射针头,分别向小鼠心肌层注射伊文思蓝(Evans Blue)和带绿色荧光蛋白的慢病毒,结果发现伊文思蓝和慢病毒能够全部存留在心肌中,而且慢病毒能够在整个心脏中稳定长期地表达绿色荧光蛋白.研究表明,改良的心肌层注射方式均一性好,手术成功率高,可用于心血管研究和基因治疗.%An improved myocardial layer injection acts as an effectual mode of adminis-tration was established. Used improved syringe,Evans Blue and green fluorescent protein (GFP) labeled lentivirus were injected in to mouse myocardial layer. Evans Blue and lentivirus were found to be stored in mouse myocardial layer, and GFP could stably ex-press in the whole heart for a long time. In conclusion, an improved myocardial layer injection may be used to cardiovascular disease(CVD)research and gene therapy research because of the good uniformity and high success rate.【期刊名称】《上海大学学报(自然科学版)》【年(卷),期】2017(023)006【总页数】6页(P835-840)【关键词】心肌层注射;基因治疗;心血管疾病【作者】王惠;赵蓉;周学中【作者单位】苏州大学附属第三医院,心血管内科,江苏常州213003;苏州大学附属第三医院,心血管内科,江苏常州213003;苏州大学附属第三医院,心血管内科,江苏常州213003【正文语种】中文【中图分类】R54心血管疾病(cardiovascular disease,CVD)严重影响着人类的健康,2013年全球因心血管疾病而死亡的人数达到1 730万,占总死亡人数的31.5%[1].随着社会经济的发展,人们生活方式的变化,心血管疾病患病人数仍在快速增长[2].近年来,随着对心血管疾病致病基因和分子基础的探索,人们已经提出了药物和基因治疗等方法缓解心血管疾病的一些症状[3].由于给药方式的选择为临床治疗和科学研究考虑的主要问题之一[4-5],本研究通过对小鼠进行心肌层注射方法的建立与优化,试图提出一种稳定有效的注射方式.1 材料与方法1.1 材料1.1.1 实验动物无特定病原体(specif i c pathogen free,SPF)级C57BL/6小鼠,雌雄均可,由常州卡文斯实验动物有限公司提供.操作依照美国国立卫生院(National Institutes of Health)的相关规定.1.1.2 注射用药物质量体积分数为1%的Evans Blue(Sigma);带GFP的慢病毒(1×109TU/mL);质量体积分数为1%的戊巴比妥钠.1.1.3 实验器材小动物呼吸机、体视显微镜、光源、加热垫、电子天平.显微外科手术器械:蚊剪、直镊、弯镊、显微尖镊、显微弯镊、高压灭菌锅、4-0手术缝合线、纱布、碘伏棉棒.胰岛素注射器,由美国BD公司提供;针头规格29 G,透明胶带.冰冻切片机、OCT包埋剂(optimal cutting temperature compound)、荧光显微镜.1.2 实验方法1.2.1 小鼠心肌层注射手术操作过程如图1所示,具体操作步骤如下.图1 小鼠心肌层注射的手术过程Fig.1 Surgical procedure of myocardial layer injection in mice第1步高压灭菌手术器械,准备手术区域,手术区域用75%的酒精擦拭消毒.第2步设置热毯温度恒定为34°C,且在手术期间保持小鼠正常的体温.设置呼吸机频率120次/min,潮气量2.0,确定小动物能够呼吸顺畅.第3步对实验小鼠进行称重,然后用质量体积分数为1%的戊巴比妥钠腹腔注射麻醉小鼠.第4步将小鼠置于34°C恒温垫,暴露小鼠颈部和胸部,用脱毛膏脱去颈部和左胸口毛发.第5步小鼠取仰卧位,用碘伏棉棒消毒手术区域3遍,镊子夹持小鼠尾巴,观察其反射程度确定小鼠是否充分麻醉.第6步在显微镜下沿直线分离覆盖气管上的颈部皮肤、肌肉及组织,暴露气管后,在声门下两个气管软骨环之间切一个小孔,插入气管插管,固定,检查胸廓的运动,确保两肺通气良好,呼吸频率为120次/min.第7步开胸.在显微镜下使用显微剪将小鼠胸骨左缘第四第五肋间位置做一横形切口,切口长约1.2 cm,逐层分离胸壁肌肉直至肋间肌暴露,使用显微镊钝性分离肋间肌肉,暴露心脏.第8步心肌注射.使用1 mL的胰岛素针吸取药物或者慢病毒稀释液一小格,即25µL,针尖垂直向下扎进心脏左心室的心室壁肌肉,扎入深度为刚没过针尖切面为标准;然后缓慢将药物推出,拔出注射器.第9步逐层缝合关闭实施手术区域.第10步对术后伤口进行碘酒消毒,若小鼠有脱水迹象,及时腹腔注射无菌生理盐水. 第11步小鼠置于热毯恢复,直到自行爬动,然后拿开热毯,将小鼠放回鼠笼.在心肌层注射过程中,针头进入的深度较难控制.进针太深会注射入心腔,药物和病毒无法进入心肌,同时心脏出血,死亡率增加;若进针太浅,药物和病毒无法注射进入心肌层,实验失败率增加.另外,注射的深浅不同,对心脏的损伤程度不一致,无法保证稳定性.基于此,采用无菌的透明胶带对针头进行处理,以此控制进针的深度并保证实验的均一性.对于不同种类的动物,比如大鼠,可以统一固定调整进针的深度.注射针头的处理和效果如图2所示.图2 改进后的小鼠心肌层注射针头Fig.2 Improved syringe of myocardial layer injection in mice1.2.2 冰冻切片与荧光拍摄冰冻切片与荧光拍摄的操作步骤如下.第1步取心脏组织.腹腔注射质量体积分数为1%的戊巴比妥钠麻醉小鼠,再用显微剪在小鼠腹部做横切口;然后由两侧垂直往上延伸直至膈肌显露,最终露出心脏;弯镊从心脏下方走行,夹住主动脉等大血管将心脏小心上提;用显微剪在弯镊下方离断血管,取出心脏并置于磷酸盐缓冲液(phosphate buあer saline,PBS)中;最后用显微镊轻轻挤压心脏,去除血液.第2步 OCT包埋剂包埋.将取出的新鲜心脏组织用OCT包埋剂浸润;然后迅速放入液氮中,快速冷却;最后置于−80°C冰箱中保存.第3步切片.预冷冰冻切片机至−20°C,然后将样品托涂上OCT包埋剂,并将样品置于其上,冰架上冷却30 min;调整角度、厚度,修片后切片,取10µm;切好的切片置于−80°C冰箱保存.第4步荧光采集.将切好的冰冻切片常温复温20 min,PBS清洗,洗净OCT包埋剂后,置于荧光显微镜下采集照片.2 结果2.1 注射药物和慢病毒效果向心肌层分别注射Evans Blue染料和慢病毒,注射完毕后,置于体视镜下观察,结果如图3和4所示.由图可以看出:注射Evans Blue染料后,注射点附近的心肌层被染成蓝色,并且向四周晕开;注射慢病毒后,注射点附近的心肌层出现白色区域.上述结果表明药物和慢病毒均可注射入心肌层,并且小鼠存活率高.图3 小鼠心肌层注射Evans Blue染料后的效果图Fig.3 Diagram of Evans Blue injection of myocardial layer injection in mice图4 小鼠心肌层注射慢病毒后的效果图Fig.4 Diagram of lentivirus injection of myocardial layer injection in mice2.2 慢病毒注射后的基因表达选取带绿色荧光蛋白(green f l uorescent protein,GFP)的慢病毒进行心肌层注射,并在注射一周后检测GFP的表达情况,结果如图5所示,其中1表示注射慢病毒的位置,标尺长度为100µm.由图可以看出,在注射点附近的不同位置均可以检测到GFP的表达,并且在其他组织中无法检测到GFP的表达(数据未显示).图5 小鼠心肌层注射慢病毒后GFP的检测结果Fig.5 Result of GFP-f l uorescent detection after myocardial layer lentivirus injection in mice3 讨论目前,心脏注射方式在一些研究中已经得到了应用,例如心内注射外泌体[6]和miRNA(microRNA)物质[7].但是心内注射会导致注射的物质随着血液进入体循环,而真正停留在心脏中的物质会偏少,影响干预效率.心肌层注射能够使绝大多数注射的物质保留在心脏中,特别是对于一些心梗区域的干预具有比较明显的优势.本研究明显地显示出注射的药物能够停留在心肌层内部.但是,心肌层注射最大的困难在于控制注射针头的深度:若注入较深,注射物质会进入心腔,并且随着血液循环而丧失,容易出现心脏出血,从而影响小鼠的存活率;若注射太浅,注射物质不易进入心脏,无法发挥注射药物的效率.本研究通过对胰岛素注射器进行改良,加入可以控制进针深度的原件,较好地解决了这一问题,提高了手术的稳定性和均一性.目前,在心血管研究中腺相关病毒(adeno-associated virus,AAV)凭借其比较高的安全性已被大家认可[8].不同的血清型对不同的组织和细胞的转染效率存在差异,例如AAV1,AAV6,AAV8和AAV9对心脏具有很好的亲和性.特别是,AAV9可通过尾静脉等常用的注射方式进行感染心脏组织[9].通过尾静脉注射,AAV9一方面大量进入心脏组织[10],另一方面还会进入脑组织[11]、肺组织[12]、肝脏组织[13]等器官,这不可避免地影响实验或者基因治疗的结果.因此,心肌层注射成为优秀的注射方案之一.本研究表明,心肌层注射可以较好地保证注射的物质停留在心脏组织并且能够长期的表达,而不影响其他器官.这为基因治疗提供了一种比较有效的注射方式.综上所述,本研究提出的心肌层注射方式为心血管研究和心血管基因治疗方案的选择提供了一定的帮助与借鉴.参考文献:[1]BENjAMIN E J,BLAHA M J,CHIUVE S E,et al.Heart disease and stroke statistics-2017 update:a report from the american heartassociation[J].Circulation,2017,135(10):e634-e1003.[2]CATUCCI M,PRETE F,BOSTICARDO M,et al.Dendritic cell functionalimprovement in a preclinical model of lentiviral-mediated gene therapy for Wiskott-Aldrich syndrome[J].Gene Therapy,2012,19(12):1150-1158.[3]TILEMANN L,ISHIkAWA K,WEBER T,et al.Gene therapy for heartfailure[J].Circulation Research,2012,110(5):777-793.[4]SU C H,WU Y J,WANG H H,et al.Nonviral gene therapy targeting cardiovascular system[J].American Journal of Physiology Heart&Circulatory Physiology,2012,303(6):629-638.[5]FERRARA K,POLLARD R,BORDEN M.Ultrasound microbubble contrast agents:fundamentals and application to gene and drug delivery[J].Annual Review of Biomedical Engineering,2007,9:415-447.[6]BEI Y,XU T,L¨U D,et al.Exer cise-induced circulating extracellular vesicles protect against cardiac ischemia-reperfusion injury[J].Basic Research in Cardiology,2017,112(4):38.[7]EULALIO A,MANO M,DAL FERRO M,et al.Functional screening identif i es miRNAs inducing cardiac regeneration[J].Nature,2012,492(7429):376-381.[8]SANTIAGORTIZ J L,SCHAFFER D V.Adeno-associated virus(AAV)vectors in cancer gene therapy[J].Journal of Controlled Release,2016,240:287-301.[9]PRASAD K M,XU Y,YANG Z,et al.Robust cardiomyocyte-specif i c gene expression following systemic injection of AAV:in vivo gene delivery follows a Poisson distribution[J].Gene Therapy,2011,18(1):43-52.[10]PLEGER S T,SHAN C,KSIENZYk J,et al.Cardiac AAV9-S100A1 gene therapy rescues postischemic heart failure in a preclinical large animal model[J].Science Translational Medicine,2011,3(92):92ra64.[11]BEY K,CIRON C,DUBREIL L,et al.Eきcient CNS targeting in adult miceby intrathecal infusion of single-stranded AAV9-GFP for gene therapy of neurological disorders[J].Gene Therapy,2017,24(5):325-332.[12]MARTINI S V,SILVA A L,FERREIRA D,et al.Tyrosine mutation in AAV9 capsid improves gene transfer to the mouse Lung[J].Cellular Physiology and Biochemistry,2016,39(2):544-553.[13]CHEN B D,HE C H,CHEN X C,et al.Targeting transgene to the heart and liver with AAV9 by diあerent promoters[J].Clininal and Experimental Pharmacology and Physiology,2015,42(10):1108-1117.。

  1. 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
  2. 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
  3. 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。

In vivo delivery of miRNAs for cancer therapy:Challenges and strategies ☆Yunching Chen a ,⁎,Dong-Yu Gao a ,Leaf Huang ba Institute of Biomedical Engineering,National Tsing Hua University,Hsinchu 30013,Taiwan,ROCbDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery,Eshelman School of Pharmacy,University of North Carolina at Chapel Hill,Chapel Hill,NC 27599,USAa b s t r a c ta r t i c l e i n f o Available online 22May 2014Keywords:miRNAGene delivery In vivo delivery Cancer therapy NanotechnologyMicroRNAs (miRNAs),small non-coding RNAs,can regulate post-transcriptional gene expressions and silence a broad set of target genes.miRNAs,aberrantly expressed in cancer cells,play an important role in modulating gene expressions,thereby regulating downstream signaling pathways and affecting cancer formation and progression.Oncogenes or tumor suppressor genes regulated by miRNAs mediate cell cycle progression,metabolism,cell death,angiogenesis,metastasis and immunosuppression in cancer.Recently,miRNAs have emerged as therapeu-tic targets or tools and biomarkers for diagnosis and therapy monitoring in cancer.Since miRNAs can regulate multiple cancer-related genes simultaneously,using miRNAs as a therapeutic approach plays an important role in cancer therapy.However,one of the major challenges of miRNA-based cancer therapy is to achieve speci fic,ef ficient and safe systemic delivery of therapeutic miRNAs in vivo .This review discusses the key challenges to the development of the carriers for miRNA-based therapy and explores current strategies to systemically deliver miRNAs to cancer without induction of toxicity.©2014Elsevier B.V.All rights reserved.Contents 1.Introduction ..............................................................1291.1.Limitations of current cancer therapies ...............................................1291.2.Advantages of miRNA-based cancer therapy ............................................1292.Mechanisms of miRNAs .........................................................1293.miRNAs in cancer ...........................................................1303.1.Cell proliferation and cell death ..................................................1313.2.Metabolism ...........................................................1313.3.Metastasis and angiogenesis ...................................................1313.4.miRNAs in cancer diagnosis and cancer therapy ...........................................1314.miRNAs:therapeutic agents or therapeutic targets?............................................1315.Pharmacokinetics and pharmacodynamics of miRNAs ...........................................1326.Current challenges in miRNA delivery ..................................................1326.1.Poor penetration of miRNAs into tumor tissues due to mechanical and biological barriers ........................1326.2.Unmodi fied miRNA antagonists and miRNA mimics are quickly degraded and cleared in the blood circulation ..............1326.3.miRNAs,as other ssRNAs or dsRNAs have the potential to induce immunotoxicity ...........................1326.4.Neurotoxicity occurs as the result of exposure to miRNAs ......................................1336.5.Poor intracellular delivery and aggregation within the endosomes of naked miRNAs result in inef ficient gene silencing (133)Advanced Drug Delivery Reviews 81(2015)128–141Abbreviations:AAVs,adeno-associated viruses;AEBP1,adipocyte enhancer-binding protein 1;AEG-1,astrocyte elevated gene-1;Ago,argonaute protein;Ago2,argonaute2protein;AMOs,anti-miRNA oligonucleotides;BBB,blood –brain-barrier;CDK6,cyclin-dependent protein kinase 6;CLL,chronic lymphocytic leukemia;CSC,cancer stem cells;DCs,dendritic cells;dsRNAs,short double strand RNAs;ECM,extracellular matrix;EGFR,epidermal growth factor receptor;EMT,epithelial –mesenchymal transition;EPR,enhanced permeability and retention;FGF,fibroblast growth factor;GBM,glioblastoma multiforme;HCC,hepatocellular carcinoma;HCV,hepatitis c virus;HDL,high-density lipoprotein;HIF-α,hypoxia-inducible factor-α;IFN,type I interferon;IL,interleukin;LAC,lung adenocarcinoma;LNA,locked nucleic acid;LPH,liposome –polycation –hyaluronic acid;mAbs,monoclonal antibodies;MCL1,myeloid cell leukemia sequence 1;MDSCs,myeloid-derived suppressor cells;miRNA,microRNA;NPs,nanoparticles;NSCLC,non-small-cell lung cancer;PEG,polyethylene glycol;PEI,polyethyleneimine;PLGA,poly(lactide-co-glycolide);pre-miRNAs,hairpin-forming miRNA precursors;pri-miRNAs,long RNA primary transcripts;PU,polyurethane;RES,reticuloendo-thelial system;RISC,RNA-induced silencing complex;scFv,single-chain variable fragment;siRNA,short interfering RNA;SLNs,solid lipid nanoparticles;SNA-NCs,spherical nucleic acid nanoparticle conjugates;SPARC,secreted protein acidic and rich in cysteine;TLRs,Toll-like receptors;TNF,tumor necrosis factor;VEGF,vascular endothelial growth factor.☆This review is part of the Advanced Drug Delivery Reviews theme issue on “miRNAs as targets for cancer treatment:Therapeutics design and delivery ”.⁎Corresponding author.E-mail address:yunching@.tw (Y.Chen)/10.1016/j.addr.2014.05.0090169-409X/©2014Elsevier B.V.All rightsreserved.Contents lists available at ScienceDirectAdvanced Drug Delivery Reviewsj o u r na l h om e p a g e :w w w.e l s e v i e r.c o m /l o c a t e /a d d r6.6.Off-target effects of miRNAs (133)6.7.Insufficient or saturated miRNA processing enzymes lead to dysfunction of therapeutic miRNAs (133)7.In vivo miRNA delivery strategies for cancer therapy (134)7.1.Local delivery of miRNAs (134)7.2.Systemic delivery of miRNAs (134)7.2.1.Modified miRNA antagonists or miRNA mimics (135)7.2.2.Viral delivery of miRNAs (135)7.2.3.Non-viral delivery of miRNAs (136)8.Future perspectives (138)Acknowledgments (138)References (138)1.IntroductionMicroRNAs(miRNAs),distinct from high-molecular-weight micro-somal RNA,are small non-coded strands of RNAs discovered in a decade [1].Many studies aid in the development of miRNA-based therapy for clinical applications.Nowadays,many of the monoclonal antibodies (mAbs)and small molecule inhibitors serve as effective cancer thera-peutics in the clinic.However,there are some limitations with regard to the specificity of inhibitors and capability of antibodies to access intracellular targets.1.1.Limitations of current cancer therapiesConventional chemotherapy,which disrupts the functions of cell organelles such as the mitochondria,cytoskeleton,inhibits the key enzyme activity to block DNA replication,mRNA transcription or trans-lation,or directly damages DNA to stop the proliferation of cancer cells and induces toxicity in cancer cells.However,the conventional cancer therapeutic agent does not target the cancer cells specifically.It also displays the toxicity in rapidly dividing normal tissues such as the bone marrow and the gastrointestinal tract,resulting in side effects [2].Therefore,the targeted therapy was developed to specifically block molecular targets regulating tumor formation and progression.The targets of small molecule inhibitors are usually overexpressed in the cancer cells and located intracellularly.For example,the tyrosine kinase inhibitor,which targets the growth factor receptors or the downstream effectors recently emerged as the systemic therapy for cancer[2–4].However,the inhibitors sometimes bind to a broad set of receptors or the downstream mediators,leading to reduced specificity and increased toxicity.Thus,monoclonal antibody-based cancer thera-py has been established and becomes one of the most efficient and safe strategies for cancer treatment[5].For example,therapeutic mAbs targeting the ERBB family including epidermal growth factor receptor(EGFR)and vascular endothelial growth factor(VEGF)showed significant therapeutic effect when treating patients with solid tumors [6,7].Recent evidences showed that EGFR-specific antibodies extended patient survival with colorectal cancer[7,8].Nevertheless,there are multiple hurdles for efficient antibody-based cancer treatment.For instance,physical properties and pharmacokinetics make it difficult for mAbs to penetrate the tumor tissue efficiently and homogeneously. Immune escape due to ineffective FcγR binding and immunosuppres-sive microenvironment leads to the reduced therapeutic efficacy[9,10].Besides,neither inhibitors nor monoclonal antibodies can success-fully treat cancer–a heterogenic disease–by suppressing a single target.Heterogeneity exists in expression between individual primary lesions,primary and metastatic lesions,and even tumor lesions before and after treatment.Particularly,it has been known tumors can develop resistant mechanisms in response to the treatment.For example,although the high-level target protein expression is detected before treatment,it may be downregulated during and after treatment as part of the resistance development.Furthermore,some cancer cells will develop the compensation mechanisms by activating other survival signaling pathways to overcome the targeted cancer treatment.For example,it has been reported that B-raf inhibitors such as vemurafenib and dabrafenib develop acquired drug resistance via hyperactivation of the PI3K/Akt pathway,leading to increased expression of adipocyte enhancer-binding protein1(AEBP1)and activation of NF-κB in melano-ma[11].To this end,the therapeutic response to the targeted agents including small molecule inhibitors and mAbs is usually partial and only causes a transient delay in tumor growth,after which most tumors continue or even accelerate their progression and metastasis[12].1.2.Advantages of miRNA-based cancer therapymiRNAs,on the other hand,can silence target genes efficiently and regulate a broad set of genes of interest simultaneously,which benefits treatment of cancer as a heterogenic disease.It has been shown that targeting a set of related oncogenic genes or pathways simultaneously triggered synergistic therapeutic effect in cancer.In spite of targeting cancer cells only,miRNAs can also target the tumor-promoting stromal cells such as endothelial cells and tumor-associatedfibroblasts to inhibit angiogenesis and tumorfibrosis,which are required during tumor formation,progression and metastasis[13–16].Moreover,miRNAs,as natural antisense nucleotides,showed reduced immune response and low toxicity when compared to plasmid DNA-based gene therapy and protein-based drug molecules.Thus,miRNAs may play a significant role in cancer therapy.As a novel therapeutic strategy,several miRNA modulators have entered the clinical trials.Locked nucleic acid(LNA)-antimir-122is thefirst drug to successfully enter Phase II trials for the treatment of hepatitis C virus(HCV)infection[17].For cancer diagnosis, miRNA-126targeting VEGF and EGFL7showed the prognostic value to provide predictive information in relation to the therapeutic outcome of anti-angiogenic agents in metastatic colorectal cancer[18].Accord-ingly,miRNA-based therapeutics may serve as promising anti-cancer drugs for the clinical application in cancer therapy if they can be efficiently and safely delivered to cancer cells.The charged miRNAs have small size and low molecular weight making them possible to be formulated into an effective delivery system and become attractive options for clinical cancer therapy development. To this end,in order to achieve effective gene silencing in cancer cells, the development of strategies for efficient in vivo delivery and escape from blood clearance,enzyme degradation and intracellular trapping, such as an endosome,is required.This review will focus on current challenges and strategies for delivery of miRNAs in vivo through local and systemic or targeted administration.The strategies employed in in vivo miRNA delivery for cancer therapy are summarized in Table1.2.Mechanisms of miRNAsThe regulation of gene expression plays an important role in mediat-ing cellular functions.Small RNA molecules,including short interfering RNAs(siRNAs)and miRNAs are effective modulators of gene expression through translation repression,chromatin remodeling or mRNA degra-dation[19].After post-transcriptional modifications,the structures of129Y.Chen et al./Advanced Drug Delivery Reviews81(2015)128–141the endogenous mature miRNAs are similar to the exogenous siRNAs. Both siRNAs and miRNAs are processed by the common enzyme—Dicer and incorporated into an active RNA-induced silencing complex (RISC).Subsequently,miRNAs and siRNAs share the same processing mechanism to achieve gene-silencing effect.However,siRNAs are exog-enous double-stranded RNAs that bind to the target mRNA sequences via perfect sequence matching.In contrast,miRNAs are endogenous single-stranded RNAs targeting multiple sequences via imperfect pairing,which leads to simultaneous suppression of multiple target genes.It creates an overwhelming advantage when applied to cancer therapy.miRNAs,referring to endogenous non-coding RNAs have recently emerged as master regulators of cancer.miRNAs can affect cancer progression through regulating the expression of target genes,which mediate cell cycle progression,metabolism,cell death,angiogenesis, metastasis and immunosuppression in cancer.As illustrated in Fig.1, miRNAs are derived from70-nucleotide hairpin-forming miRNA pre-cursors(pre-miRNAs)after long RNA primary transcripts(pri-miRNAs) are cleaved by the ribonuclease III termed DROSHA in the nucleus.Pre-miRNAs are subsequently transported to the cytoplasm by exportin-5 and further converted into double-stranded18~25nt RNAs by Dicer. Mature miRNAs,the short double strand RNAs(dsRNAs),are incorpo-rated into the RISC complex composed of Dicer,many associated proteins,and target mRNAs carrying complementary sequences.One of the strands of mature miRNAs,known as the guide strand,binds to Argonaute(Ago)protein in the RISC complex,while the passenger strand is degraded.By the imperfect base pairing between the guide strands of miRNAs and the3′or5′untranslated region of the target mRNAs,miRNAs are able to regulate tens to hundreds of mRNAs. Despite only the imperfect base pairing between miRNAs and the3′or 5′untranslated region of the target mRNAs is required for miRNAs to regulate the expressions of target mRNAs,the“seed”region (nucleotides2~7from the5′end of the miRNA)of the miRNA still have to be perfectly matched with the complementary mRNA sequence [20].The miRNA–mRNA interaction suppresses the expression levels of the target genes through mRNA cleavage or translational regression.The gene suppression mechanism depends on the degree of complementarity sites between the miRNAs and mRNAs[21,22]. Extensive base pairing is required for Argonaute2(Ago2)endonuclease-mediated mRNA cleavage,while moderate base pairing is adequate to achieve translational inhibition[21–23].Interestingly,some miRNAs generated in the cytoplasm cannot only affect the function of the cell that produces the miRNAs,but also be released into the bloodstream and taken up by other cells to regulate gene expression of the distant target cells.These endogenous miRNAs released into plasma are packaged in microparticles(exosomes, microvesicles,and apoptotic bodies)or bound to RNA-binding proteins such as Ago2or lipoprotein complexes such as high-density lipoprotein (HDL)to maintain their stabilities[24–26].It was found that the cell-free miRNAs in plasma could also be detected and expressed in platelets, erythrocytes,and nucleated blood cells[27,28].For example,of79 tumor-specific circulating miRNAs,58%(46of79)are highly expressed in one or more blood cell types such as myeloid(e.g.,miR-223,miR-197, miR-574-3p,and let-7a),lymphoid blood cells(e.g.,miR-150),and red blood cells(e.g.,miR-486-5p,miR-451,miR-92a,and miR-16)[29]. Consequently,miRNAs can target various gene expressions,contribute cell-to-cell communication and regulate the key cell signaling pathways to maintain regular functions in normal cells.Thus,abnormal miRNA expressions may participate in cancer formation and progression.3.miRNAs in cancerMisexpression or dysfunction of miRNAs is associated with tumor formation and progression via manipulating the oncogenic pathways that influence the processes in tumor progression,such as cell cycle regulation,apoptosis,senescence,metabolism,angiogenesis and metastasis.Table1Summary of studies using miRNA for cancer therapy in vivo.Vehicle Targeted miRNA Antagonistor mimicsModel Effect ReferenceLocal DeliveryCholesterol-conjugated2′-O-methyl-modifiedmiR-375Mimics Hepatoma xenograft Inhibition of tumor growth[94] Lentiviral vector let-7Mimics Non-small-cell lung cancer(NSCLC)Inhibition of the growth of K-ras-dependent lung tumors[95]Polyethyleneimine(PEI)miR-145Mimics Colon carcinoma xenograft Induction of apoptosis,inhibition of tumor growth anddownregulation of the oncogenic kinase Pim-1[96]miR-33aSystemic deliverySeed-targeting tiny LNAs miR-21Antagonist Breast cancer Repression of the miR-21function in tumor[106] Cationic liposomes miR-143Mimics Colorectal carcinoma Inhibition of tumor growth[109] Lentiviral vectors miR-15a/16Mimics Chronic lymphocytic leukemia Restoration of miR-15a/16expression and inhibition oftumor cell proliferation[110]miR-494Antagonist Breast cancer Inhibition of tumor growth and metastasis[111] Adeno-associated viruses(AAVs)miR-26a Mimics Hepatocellular carcinoma Inhibition of tumor cell proliferation and induction of apoptosis[112] Silica nanoparticles miR-34a Mimics Neuroblastoma Induction of apoptosis,reduction in vascular density oftumors and inhibition of tumor growth[101]PEGylated-PLGA miR-21Antagonists Breast cancer Efficient delivery of antagomiR-21and prolonged release[125] PLGA-penetratin miR-155Antagonists Lymphoma Induction of apoptosis and reduction of tumor growth[126] PEI miR-145Mimics Colorectal carcinoma Reduction of tumor cell proliferation and tumor growth[130] miR-33aLNP-DP1miR-122Mimics Hepatocellular carcinoma Inhibition of angiogenesis and tumor growth[132] Cationic DOTMA lipoplexes miR-133b Mimics NSCLC Increased expression of miR-133b and downregulation ofprosurvival gene MCL-1[133] miR-29b Mimics NSCLC Reduced expression of the key target oncogenes andinhibition of tumor growth[134] Neutral lipid miR-34a Mimics NSCLC Inhibition of tumor growth[136] miR-34a Mimics NSCLC Inhibition of tumor cell proliferation and induction of apoptosis[137]let-7LPH-PEG-GC4miR-34a Mimics Lung cancer Reduction of tumor growth,induction of apoptosis,inhibition of survivin expression and downregulationof MAPK pathway [102]130Y.Chen et al./Advanced Drug Delivery Reviews81(2015)128–1413.1.Cell proliferation and cell deathFive groups of miRNAs,including the miR-15a/16cluster,the miR-17/20cluster,the miR-221/222cluster,and the let-7and miR-34 families,can target cell cycle regulators to control cell cycle checkpoints and progression[30].Dysfunction of the cell cycle-related miRNAs increases cell proliferation,leading to tumor growth promotion.In contrast,recent study has highlighted miRNAs as anti-apoptotic or pro-apoptotic regulators by targeting various mRNAs associated with key apoptotic pathways in cancer.For instance,miRNA-221/222cluster targeting p27,PTEN,and PUMA,miRNA-128targeting BAX and miRNA-17/19cluster targeting BIM act as anti-apoptotic miRNAs to maintain survival of cancer cells and contribute to drug resistance[31–33].On the other hand,pro-apoptotic miRNAs15a/b and16targeting BCL2 family serve as anti-cancer guardians[34].3.2.MetabolismThe miRNA expression profiles associated with metabolism are distinct from normal cells.miRNAs play as key regulators of metabolism in cancer cells to increase nutrient uptake and the accumulation of materials for controlling metabolicflux[35].For example,miRNAs such as miR-133,miR-138and miR-150targeting glucose transporter, miR-33a/b targeting metabolic enzymes to regulate fatty acid metabo-lism and AMPK pathway and miR-29b targeting amino acid catabolism are involved in modulating cancer cell metabolism and biogenesis to support tumor growth and proliferation[36–39].3.3.Metastasis and angiogenesismiRNAs have impact on both intrinsic signaling pathways of cancer cells and interactions between cancer cells and tumor stroma to regulate invasion and metastasis.For instance,miR-200family and miR-205are downregulated in various tumors to promote epithelial–mesenchymal transition(EMT)progression and facilitate cancer cell invasion[40].miR-29b upregulated in breast cancer cells suppresses the expression of MMP2and MMP9and consequently triggers extracellular matrix(ECM)remodeling to facilitate cancer cell migration and local invasion[40].Besides,angiogenesis is required for supporting the formation of both primary and metastatic lesions. miR-424triggered by hypoxia stabilizes hypoxia-inducible factor-α(HIF-α)and enhances angiogenesis in tumor microenvironment[41]. miR-503suppresses the expression of angiogenic factors likefibroblast growth factor(FGF)2and VEGF-A.miR-503is downregulated in hepatocellular carcinoma(HCC),resulting in increased angiogenesis and tumorigenesis[42].3.4.miRNAs in cancer diagnosis and cancer therapyAs we summarized above,the aberrant expressions of miRNAs are associated with tumor formation,progression and metastasis. Moreover,the abnormal expressions of miRNAs are also correlated with the resistant mechanisms to chemotherapy.Thus,miRNAs can serve as both diagnostic and prognostic biomarkers in cancer[43].For example,miR-155overexpression and let-7a downregulation are associated with poor disease outcome in lung cancer[44].For prognos-tic applications,overexpression of miR-221and miR-222are associated with poor therapeutic outcome of anti-estrogenic therapies such as Tamoxifen and Fulvestran,while tumor suppressor miR-205is responsible for enhanced therapeutic effect of tyrosine kinase inhibitors [45–47].Many reports have demonstrated the significance of micro-RNAs as diagnostic and prognostic biomarkers.Besides,miRNAs can also serve as either therapeutic agents or therapeutic targets in cancer. In the following sections,we will further focus on the therapeutic applications of miRNAs.4.miRNAs:therapeutic agents or therapeutic targets?According to the functions of miRNAs,cancer types and stages, both antagonists and mimics are developed as miRNA-based therapeutic strategies to achieve tumor regression[48].miRNA antago-nists–single-stranded oligonucleotides with miRNA complementary sequences–are designed to interrupt the miRNA processing as well as RISC assembly,and result in increased expression of thetumorFig.1.Schematic representation of the microRNA generation and silencing mechanisms.Hairpin-forming pre-miRNAs are generated by pri-miRNAs,which is cleaved by ter,pre-miRNAs are transported into the cytoplasm by exportin-5and further converted into double-stranded mature miRNAs by Dicer.Mature miRNAs are incorporated into the RISC complex, unwound and annealed to the target mRNAs carrying complementary sequences.miRNAs are able to regulate tens to hundreds of mRNAs via the imperfect base pairing between miRNAs and the3′or5′untranslated region of the target mRNAs.The miRNA-mRNA interaction silences the target genes through mRNA cleavage or translational inhibition.131Y.Chen et al./Advanced Drug Delivery Reviews81(2015)128–141suppressor genes.For example,miR-21,overexpressed in various tumor types,downregulates many tumor suppressor genes regulating cell proliferation,cell death,metastasis and chemoresistance.miR-21 antagonist reverses EMT phenotype and blocks angiogenesis in breast cancer through inactivation of AKT and MAPK pathways[49,50].The targeted endogenous miRNA can also serve as a noninvasive biomarker for early cancer diagnosis,prediction of response to miRNA antagonist therapy and therapy monitoring.By contrast with miRNA antagonists,miRNA mimics,known as miRNA replacement therapy,play an opposite role in regulating the expression of target genes.Genomic loss of tumor suppressor miRNAs can be restored by miRNA mimics,which behave like endogenous miRNAs.Moreover,miRNA mimics cannot only provide obvious bene-fits to those cancer cells with low tumor suppressor miRNA expression levels,but also show therapeutic benefits in cancer with normal miRNA expression levels.For example,the miR-34family is dysregulated in different cancer types including several epithelial tumors,melanomas, neuroblastomas,leukemias and sarcomas[51].miR-34serves as a downstream effector of p53pathway,which is defective in about half of human cancers and plays an important role in the suppression of tumor development[52].Therefore,miR-34is referred as a potential tumor suppressor and a possible therapeutic target.Interestingly, delivery of miR-34mimics to cancer cells with both reduced and normal expression levels of miR-34showed growth inhibitory effect.Therefore, miRNA mimics could be a promising treatment for various types and stages of cancer diseases.During miRNA processing,double-stranded miRNAs are loaded onto the RISC complex and one strand of the miRNAs,the passenger strand,is cleaved by Ago2.Furthermore,the other RNA strand,the guide strand, remains and matches the complementary sequences of the target mRNAs,leading to mRNA cleavage or translational repression.Thus, the information of the miRNA guide strands is more important for designing miRNA antagonists or miRNA mimics.Both miRNA antagonists and miRNA mimics are low molecular weight oligonucleotides and thus easier to deliver into the target cells compared with large viral vectors or plasmids normally used for gene therapy.However,the lack of correlation between in vitro and in vivo efficacies was observed due to inefficient in vivo delivery of miRNAs. Like other therapeutic oligonucleotides,the delivery of the miRNA antagonists or mimics as cancer therapies encounter several barriers such as poor bioavailability,limited tissue permeability and payload instability.5.Pharmacokinetics and pharmacodynamics of miRNAsmiRNA antagonists or miRNA mimics,the poly-anionic molecules with low molecular weights,are highly water-soluble and suitable for intravenous and subcutaneous injections.After intravenous admin-istration,the plasma levels of miRNA antagonists or miRNA mimics reduce quickly.They further distribute broadly but later accumulate mostly in the liver and kidney.Tissue concentrations of miRNA antago-nists or miRNA mimics in the brain,heart and lung decline rapidly after systemic injection.Nevertheless,tissue concentrations of miRNA-based therapeutics in the liver and kidney remain high and sustained levels up to24h after injection[53].The modified miRNAs show distinct pharmacokinetics from unmodified ones.As unmodified miRNAs, plasma levels of modified miRNA antagonists or miRNA mimics are reduced within hours and accumulated into tissues.However,after entering into cells,the modified miRNAs remain stable that their clearance rate in tissues is reduced,leading to prolonged therapeutic benefit.The duration of the pharmacological effects(or pharmacodynamics) of miRNA antagonists or miRNA mimics is determined by their retention in the target tissues.On the other hand,the onset of their pharmacological effects is often delayed because of the time delay between internalization of miRNA antagonists/miRNA mimics and regulation of the target proteins.Furthermore,the fact that miRNA-based therapeutics may indirectly mediate the diseased phenotype further delays the pharmacological effect.For example,both miR-122 and mi-208antagonists have delayed effect on their target cholesterol andβ-MHC respectively and cause a postponed change in the disease phenotype[54–56].To this end,there are still many challenges remain-ing in developing miRNAs as effective therapeutic agents in cancer.6.Current challenges in miRNA deliverySeveral problems encountered in clinical development of miRNA delivery limit the application of miRNAs as a therapeutic option to treat cancer[57–60](Fig.2).6.1.Poor penetration of miRNAs into tumor tissues due to mechanical and biological barriersThe major challenge of miRNA delivery into cancer is to successfully deliver miRNA antagonists or miRNA mimics to the target tumor tissue with efficient penetration of cargos into the tumor.The leaky structure and compression of abnormal tumor vessels lead to poor blood perfusion,which reduces the delivery efficacy of naked miRNAs[61] (Fig.2).In addition,the slowdown of miRNA diffusion in solid tumors caused by higher interstitialfluid pressure holds the hurdle of miRNA delivery in cancer therapy[62].The complex extracellular matrix (ECM)also plays an important role to hinder miRNAs from reaching the cancer cells(Fig.2).For example,thefibrotic microenvironment of pancreatic cancer results in poor diffusion of therapeutic drugs[63, 64].The nonmalignant cells in the tumor microenvironment also appear to be an important extracellular barrier.For instance,tumor-associated macrophages,neutrophils and monocytes can nonspecifically uptake and trap miRNAs encapsulated in the delivery system[65].Besides, the blood–brain-barrier(BBB)represents a problem to the miRNA therapy involving cancer in central nervous system.Tight junctions between the brain endothelial cells reduce miRNA diffusion and delivery into brain tumors[66].6.2.Unmodified miRNA antagonists and miRNA mimics are quickly degraded and cleared in the blood circulationThe other challenge that remains in miRNA delivery is to maintain the stability and integrity of miRNAs in circulation.Naked miRNAs are degraded within seconds by the abundant nucleases such as serum RNase A-type nucleases in the blood(Fig.2)[67].In addition,naked miRNAs are cleared rapidly via renal excretion,leading to a short half-life in systemic circulation[68].miRNAs carried by nanoparticles larger than100nm in diameter increase the reticuloendothelial system(RES) clearance in the liver,spleen,lung and bone marrow,which results in non-specific uptake by innate immune cells such as monocytes and macrophages(Fig.2)[69].6.3.miRNAs,as other ssRNAs or dsRNAs have the potential toinduce immunotoxicitySystemic miRNA delivery,like other types of nucleic acid,activates innate immune system leading to unexpected toxicities and significant undesirable side effects.Systemic administration of miRNA duplexes can trigger secretion of inflammatory cytokines and type I interferons (IFNs)through Toll-like receptors(TLRs)(Fig.2).TLRs3,7and8are activated by single or double-stranded RNAs(dsRNAs)to drive innate and adaptive immune responses.These TLRs sensing dsRNA molecules in cellular endosomal and lysosomal compartments stimulate the type I interferon(IFN)pathway and trigger cytokine production according to the structure,sequence,and the delivery system[70,71].IFN induced by the activation of TLRs further upregulates the expressions of TLRs and primes the surrounding immune cells such as monocytes,dendritic132Y.Chen et al./Advanced Drug Delivery Reviews81(2015)128–141。

相关文档
最新文档