EPR, enhanced permeability and retention

EPR,增强渗透性和保留
  • 文章类型: Journal Article
    纳米颗粒药物递送系统(Nano-DDS)已经成为抗癌药物递送障碍的可能解决方案。然而,临床结果和翻译受到几个缺点的限制,如低药物负载,药物过早泄漏和载体相关毒性。最近,纯药物纳米组件(PDNAs),通过纯药物分子的自组装或共组装制造,引起了相当大的关注。他们的简便和可重复的制备技术有助于消除纳米药物的瓶颈,包括质量控制,扩大生产和临床翻译。既是承运人又是货物,无载体的PDNA具有超高或甚至100%的载药量。此外,基于PDNA的联合疗法可能解决癌症治疗中最棘手的问题,如肿瘤转移和耐药。在本次审查中,概述了PDNA用于癌症治疗的最新进展。首先,PDNA根据药物分子的组成进行分类,并对装配机理进行了讨论。此外,总结了用于联合治疗的PDNA的共同递送,特别关注治疗结果的改善。最后,PDNA用于有效癌症治疗的未来前景和挑战受到关注。
    Nanoparticulate drug delivery systems (Nano-DDSs) have emerged as possible solution to the obstacles of anticancer drug delivery. However, the clinical outcomes and translation are restricted by several drawbacks, such as low drug loading, premature drug leakage and carrier-related toxicity. Recently, pure drug nano-assemblies (PDNAs), fabricated by the self-assembly or co-assembly of pure drug molecules, have attracted considerable attention. Their facile and reproducible preparation technique helps to remove the bottleneck of nanomedicines including quality control, scale-up production and clinical translation. Acting as both carriers and cargos, the carrier-free PDNAs have an ultra-high or even 100% drug loading. In addition, combination therapies based on PDNAs could possibly address the most intractable problems in cancer treatment, such as tumor metastasis and drug resistance. In the present review, the latest development of PDNAs for cancer treatment is overviewed. First, PDNAs are classified according to the composition of drug molecules, and the assembly mechanisms are discussed. Furthermore, the co-delivery of PDNAs for combination therapies is summarized, with special focus on the improvement of therapeutic outcomes. Finally, future prospects and challenges of PDNAs for efficient cancer therapy are spotlighted.
    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
    Drug transportation is impeded by various barriers in the hypoxic solid tumor, resulting in compromised anticancer efficacy. Herein, a solid lipid monostearin (MS)-coated CaO2/MnO2 nanocarrier was designed to optimize doxorubicin (DOX) transportation comprehensively for chemotherapy enhancement. The MS shell of nanoparticles could be destroyed selectively by highly-expressed lipase within cancer cells, exposing water-sensitive cores to release DOX and produce O2. After the cancer cell death, the core-exposed nanoparticles could be further liberated and continue to react with water in the tumor extracellular matrix (ECM) and thoroughly release O2 and DOX, which exhibited cytotoxicity to neighboring cells. Small DOX molecules could readily diffuse through ECM, in which the collagen deposition was decreased by O2-mediated hypoxia-inducible factor-1 inhibition, leading to synergistically improved drug penetration. Concurrently, DOX-efflux-associated P-glycoprotein was also inhibited by O2, prolonging drug retention in cancer cells. Overall, the DOX transporting processes from nanoparticles to deep tumor cells including drug release, penetration, and retention were optimized comprehensively, which significantly boosted antitumor benefits.
    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
    炎症性肠病(IBD)是一种慢性肠道疾病,临床表现痛苦,癌变风险高。目前没有治疗IBD的治疗方法,大力提倡开发有效的疗法。已经广泛研究了药物递送系统,以通过由炎症引起的增强的渗透性和保留(EPR)效应将治疗剂传递到发炎的结肠部位。然而,由于非靶向药物释放,该药物仍然无法达到仅利用EPR效应的有效浓度值,并且在发炎区域显示出更好的治疗效果.大量研究表明,IBD发生时,一些特异性受体和细胞粘附分子在结肠内皮和/或免疫细胞表面高表达,靶向此类受体和细胞粘附分子的配体修饰的药物递送系统可以特异性地将药物递送到发炎部位并获得巨大的疗效。本文介绍了结肠炎症部位过表达的受体和细胞粘附分子,并回顾了相关配体功能化的药物传递系统。最后,提出了该领域的挑战和未来方向,以推进用于治疗IBD的受体介导的靶向药物递送系统的开发。
    Inflammatory bowel disease (IBD) is a chronic intestinal disease with painful clinical manifestations and high risks of cancerization. With no curative therapy for IBD at present, the development of effective therapeutics is highly advocated. Drug delivery systems have been extensively studied to transmit therapeutics to inflamed colon sites through the enhanced permeability and retention (EPR) effect caused by the inflammation. However, the drug still could not achieve effective concentration value that merely utilized on EPR effect and display better therapeutic efficacy in the inflamed region because of nontargeted drug release. Substantial researches have shown that some specific receptors and cell adhesion molecules highly expresses on the surface of colonic endothelial and/or immune cells when IBD occurs, ligand-modified drug delivery systems targeting such receptors and cell adhesion molecules can specifically deliver drug into inflamed sites and obtain great curative effects. This review introduces the overexpressed receptors and cell adhesion molecules in inflamed colon sites and retrospects the drug delivery systems functionalized by related ligands. Finally, challenges and future directions in this field are presented to advance the development of the receptor-mediated targeted drug delivery systems for the therapy of IBD.
    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
    血脑屏障(BBB)严格控制血液和大脑之间的物质交换,并严重限制全身给药的大脑渗透,导致脑部疾病的药物治疗无效。然而,在脑部疾病的发生和发展过程中,BBB改变不可避免地演变。在这次审查中,我们专注于基于BBB进化设计的纳米级脑靶向药物递送策略以及包括阿尔茨海默病在内的各种脑疾病的相关应用,帕金森病,癫痫,中风,创伤性脑损伤和脑肿瘤。优化BBB杂交和非全身给药途径的小分子研究进展(例如,本综述不包括BBB旁路的鼻内治疗)。
    Blood-brain barrier (BBB) strictly controls matter exchange between blood and brain, and severely limits brain penetration of systemically administered drugs, resulting in ineffective drug therapy of brain diseases. However, during the onset and progression of brain diseases, BBB alterations evolve inevitably. In this review, we focus on nanoscale brain-targeting drug delivery strategies designed based on BBB evolutions and related applications in various brain diseases including Alzheimer\'s disease, Parkinson\'s disease, epilepsy, stroke, traumatic brain injury and brain tumor. The advances on optimization of small molecules for BBB crossing and non-systemic administration routes (e.g., intranasal treatment) for BBB bypassing are not included in this review.
    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
    免疫疗法是癌症治疗的快速发展领域,因为其具有比传统疗法更高的特异性和具有更大功效的潜力。通过给药进行免疫细胞调节,蛋白质,和细胞可以通过在免疫抑制肿瘤存在下可能被抑制的途径增强抗肿瘤反应。磁系统为提高免疫疗法的性能提供了几个优点。包括增加对运输的时空控制,释放,以及体内免疫调节药物的剂量,导致减少的脱靶效应和提高疗效。与光和pH等刺激药物释放的替代方法相比,磁系统使几种不同的方法编程免疫反应。首先,我们讨论了磁性热疗如何刺激免疫细胞并引发热响应性药物释放。第二,我们总结了药物载体的磁性靶向递送如何增加药物在靶位点的积累。第三,我们回顾了生物材料如何经历磁驱动的结构变化,以实现封装药物的远程释放。第四,我们描述了使用磁性颗粒与细胞受体的靶向相互作用以促进抗肿瘤活性。最后,我们讨论这些系统的翻译考虑因素,如毒性,临床相容性,以及改善癌症治疗的未来机会。
    Immunotherapy is a rapidly developing area of cancer treatment due to its higher specificity and potential for greater efficacy than traditional therapies. Immune cell modulation through the administration of drugs, proteins, and cells can enhance antitumoral responses through pathways that may be otherwise inhibited in the presence of immunosuppressive tumors. Magnetic systems offer several advantages for improving the performance of immunotherapies, including increased spatiotemporal control over transport, release, and dosing of immunomodulatory drugs within the body, resulting in reduced off-target effects and improved efficacy. Compared to alternative methods for stimulating drug release such as light and pH, magnetic systems enable several distinct methods for programming immune responses. First, we discuss how magnetic hyperthermia can stimulate immune cells and trigger thermoresponsive drug release. Second, we summarize how magnetically targeted delivery of drug carriers can increase the accumulation of drugs in target sites. Third, we review how biomaterials can undergo magnetically driven structural changes to enable remote release of encapsulated drugs. Fourth, we describe the use of magnetic particles for targeted interactions with cellular receptors for promoting antitumor activity. Finally, we discuss translational considerations of these systems, such as toxicity, clinical compatibility, and future opportunities for improving cancer treatment.
    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)

  • 文章类型: Journal Article
    急性呼吸窘迫综合征(ARDS)的特征是严重的炎症和肺部气血屏障的破坏,导致不可逆和实质性的呼吸功能损害。2019年冠状病毒病(COVID-19)患者遇到了ARDS的高风险,强调开发有效疗法的紧迫性。然而,由于药代动力学不佳,仍然缺乏治疗ARDS的适当药物,非特异性副作用,无法克服肺屏障,和异质性管理不足。在ARDS的病理环境中增加的肺通透性可能有助于纳米颗粒介导的被动靶向递送。纳米医学在解决ARDS药物治疗困境方面表现出独特的优势,可以解决传统抗炎或抗氧化药物治疗的缺点和局限性。通过被动,活跃,或物理化学靶向,纳米载体可以与肺上皮/内皮和炎症细胞相互作用,以逆转异常变化并恢复肺环境的稳态,从而显示出良好的治疗活性和降低的毒性。本文综述了纳米医学在临床前ARDS治疗中的最新应用,强调肺部炎症的靶向治疗策略,展示了创新的药物输送系统,并为加强纳米医学治疗的治疗效果提供了启示。
    Acute respiratory distress syndrome (ARDS) is characterized by the severe inflammation and destruction of the lung air-blood barrier, leading to irreversible and substantial respiratory function damage. Patients with coronavirus disease 2019 (COVID-19) have been encountered with a high risk of ARDS, underscoring the urgency for exploiting effective therapy. However, proper medications for ARDS are still lacking due to poor pharmacokinetics, non-specific side effects, inability to surmount pulmonary barrier, and inadequate management of heterogeneity. The increased lung permeability in the pathological environment of ARDS may contribute to nanoparticle-mediated passive targeting delivery. Nanomedicine has demonstrated unique advantages in solving the dilemma of ARDS drug therapy, which can address the shortcomings and limitations of traditional anti-inflammatory or antioxidant drug treatment. Through passive, active, or physicochemical targeting, nanocarriers can interact with lung epithelium/endothelium and inflammatory cells to reverse abnormal changes and restore homeostasis of the pulmonary environment, thereby showing good therapeutic activity and reduced toxicity. This article reviews the latest applications of nanomedicine in pre-clinical ARDS therapy, highlights the strategies for targeted treatment of lung inflammation, presents the innovative drug delivery systems, and provides inspiration for strengthening the therapeutic effect of nanomedicine-based treatment.
    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
    脂质体作为药物递送载体在临床上取得了令人瞩目的成就。脂质体产品主要受益于远程药物装载技术,该技术在两亲性和/或可电离药物中取得成功,但对于非电离和水溶性差的治疗剂似乎不切实际,从而阻碍了广泛的有希望的药物搭便车脂质体用于疾病治疗。在这项研究中,通过简单的一步合成设计了一系列弱酸药物衍生物,可以通过pH梯度法远程加载到脂质体中。选择卡巴他赛(CTX)弱酸衍生物来评估其安全性,药效学,和药代动力学。CTX弱酸衍生物脂质体在安全性方面优于Jevtana®,包括全身毒性,血液学毒性,和潜在的中枢神经毒性。具体来说,研究表明,脂质体具有减弱CTX对皮质和海马神经元潜在毒性的能力.由于更高的安全性和升高的耐受剂量,在前列腺癌和转移癌治疗中实现了CTX弱酸衍生物负载脂质体的显著优势。
    Liposomes have made remarkable achievements as drug delivery vehicles in the clinic. Liposomal products mostly benefited from remote drug loading techniques that succeeded in amphipathic and/or ionizable drugs, but seemed impracticable for nonionizable and poorly water-soluble therapeutic agents, thereby impeding extensive promising drugs to hitchhike liposomal vehicles for disease therapy. In this study, a series of weak acid drug derivatives were designed by a simplistic one step synthesis, which could be remotely loaded into liposomes by pH gradient method. Cabazitaxel (CTX) weak acid derivatives were selected to evaluate regarding its safety profiles, pharmacodynamics, and pharmacokinetics. CTX weak acid derivative liposomes were superior to Jevtana® in terms of safety profiles, including systemic toxicity, hematological toxicity, and potential central nerve toxicity. Specifically, it was demonstrated that liposomes had capacity to weaken potential toxicity of CTX on cortex and hippocampus neurons. Significant advantages of CTX weak acid derivative-loaded liposomes were achieved in prostate cancer and metastatic cancer therapy resulting from higher safety and elevated tolerated doses.
    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
    这里,在脂质体纳米平台中整合了eviodiamine(EVO)和光敏剂吲哚菁绿(ICG),用于口腔鳞状细胞癌(OSCC)的非侵入性诊断成像和联合治疗.EVO,作为从中药中提取的活性成分,不仅具有抗肿瘤化疗剂的功能,而且能够进行68Ga螯合,因此作为正电子发射断层扫描/计算机断层扫描(PET/CT)成像的造影剂。此外,EVO可以表现出过氧化物酶样的催化活性,将内源性肿瘤H2O2转化为细胞毒性活性氧(ROS),使化学催化疗法超越了众所周知的EVO化疗效果。体外和体内实验证明,由光学成像和PET/CT成像引导,研究表明,治疗性脂质体通过光动力疗法联合化学动力化疗对原位舌癌有明显的抑制作用。
    Here, evodiamine (EVO) and the photosensitizer indocyanine green (ICG) were integrated into a liposomal nanoplatform for noninvasive diagnostic imaging and combinatorial therapy against oral squamous cell carcinoma (OSCC). EVO, as an active component extracted from traditional Chinese medicine, not only functioned as an antitumor chemotherapeutic agent but was also capable of 68Ga-chelation, thus working as a contrast agent for positron emission tomography/computed tomography (PET/CT) imaging. Moreover, EVO could exhibit peroxidase-like catalytic activity, converting endogenous tumor H2O2 into cytotoxic reactive oxygen species (ROS), enabling Chemo catalytic therapy beyond the well-known chemotherapy effect of EVO. As proven by in vitro and in vivo experiments, guided by optical imaging and PET/CT imaging, we show that the theragnostic liposomes have a significant inhibiting effect on in situ tongue tumor through photodynamic therapy combined with chemodynamic chemotherapy.
    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