Blood-brain barrier (BBB)

血脑屏障 ( BBB )
  • 文章类型: Journal Article
    苯扎贝特(BEZ)在不同类型的神经系统疾病中显示出广泛的神经保护作用。然而,其在创伤性脑损伤(TBI)中的药理功能尚不清楚。在目前的研究中,在小鼠中构建TBI模型以检查BEZ的潜在有益作用。在TBI之后,每天用BEZ或媒介物溶液对小鼠进行节食。电机功能,学习和记忆,脑水肿,血管炎症因子,血脑屏障(BBB)的完整性,并评估了紧密连接带闭塞1(ZO-1)的表达。研究结果表明,在TBI之后,BEZ治疗显著促进运动功能和认知功能缺损的恢复。此外,BEZ通过降低脑含水量来减轻脑水肿。我们还发现,BEZ的给药通过抑制ICAM-1,VCAM-1和E-选择素的表达来减轻脑血管炎症。值得注意的是,BEZ通过恢复紧密连接(TJ)蛋白ZO-1的表达改善了TBI小鼠受损的BBB完整性。进一步的体外实验表明,用BEZ处理可以防止内皮通透性的恶化,并恢复TBI暴露的脑bEnd.3细胞中跨上皮电阻(TEER)的降低以及ZO-1的表达。机械上,我们证明BEZ的保护作用是由AMPK介导的。基于这些发现,我们得出的结论是,BEZ可以改善TBI引起的BBB损伤,可以考虑将其用于TBI的治疗或管理。
    Bezafibrate (BEZ) has displayed a wide range of neuroprotective effects in different types of neurological diseases. However, its pharmacological function in traumatic brain injury (TBI) is still unknown. In the current study, a TBI model was constructed in mice to examine the potential beneficial roles of BEZ. After TBI, mice were daily dieted with BEZ or vehicle solution. The motor function, learning and memory, brain edema, vascular inflammatory factors, the integrity of the blood-brain barrier (BBB), and the expression of the tight junction zona occludens 1 (ZO-1) were assessed. The findings demonstrate that after TBI, BEZ treatment significantly promoted the recovery of motor function and cognitive function deficits. Moreover, BEZ attenuated brain edema by reducing the levels of brain water content. We also found that administration of BEZ alleviated cerebral vascular pro-inflammation by suppressing the expression of ICAM-1, VCAM-1, and E-selectin. Notably, BEZ improved the impaired BBB integrity in TBI mice by restoring the expression of the tight junction (TJ) protein ZO-1. Further in vitro experiments show that treatment with BEZ prevented the aggravation of endothelial permeability and restored the reduction of trans-epithelial electrical resistance (TEER) as well as the expression of ZO-1 in TBI-exposed brain bEnd.3 cells. Mechanistically, we prove that the protective effects of BEZ are mediated by AMPK. Based on these findings, we conclude that BEZ improves TBI-induced BBB injury and it might be considered for the treatment or management of TBI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    这次审查,由国际化学物质研究学会(ISSX)新研究者牵头的关于药物-药物相互作用(DDIs)的特刊的一部分,探索药物转运蛋白在吸收中的关键作用,处置,以及DDI背景下的许可。在过去的二十年里,在理解这些转运蛋白的临床相关性方面取得了重大进展.总结了有关影响药物处置和开发的关键摄取和外排转运蛋白的最新知识。FDA的监管指南,EMA,详细讨论了为潜在转运蛋白介导的DDI评估提供信息的PMDA。回顾了评估潜在DDI的临床前和临床试验方法。重点是基于生理的药代动力学(PBPK)建模的实用性。这包括应用相对丰度和表达因子来使用临床前数据预测人体药代动力学(PK),整合最新的监管指引。评估特殊人群中转运蛋白介导的DDI的考虑因素,包括儿科,肝,和肾损害组,提供。此外,探讨了血脑屏障(BBB)转运蛋白对CNS相关药物处置的影响。加强对药物转运体及其在药物处置和毒性中的作用的认识,可以提高疗效,减少不良反应。持续的研究对于弥合知识方面的剩余差距至关重要,特别是与细胞色素P450(CYP)酶相比。
    This review, part of a special issue on drug-drug interactions (DDIs) spearheaded by the International Society for the Study of Xenobiotics (ISSX) New Investigators, explores the critical role of drug transporters in absorption, disposition, and clearance in the context of DDIs. Over the past two decades, significant advances have been made in understanding the clinical relevance of these transporters. Current knowledge on key uptake and efflux transporters that affect drug disposition and development is summarized. Regulatory guidelines from the FDA, EMA, and PMDA that inform the evaluation of potential transporter-mediated DDIs are discussed in detail. Methodologies for preclinical and clinical testing to assess potential DDIs are reviewed, with an emphasis on the utility of physiologically based pharmacokinetic (PBPK) modeling. This includes the application of relative abundance and expression factors to predict human pharmacokinetics (PK) using preclinical data, integrating the latest regulatory guidelines. Considerations for assessing transporter-mediated DDIs in special populations, including pediatric, hepatic, and renal impairment groups, are provided. Additionally, the impact of transporters at the blood-brain barrier (BBB) on the disposition of CNS-related drugs is explored. Enhancing the understanding of drug transporters and their role in drug disposition and toxicity can improve efficacy and reduce adverse effects. Continued research is essential to bridge remaining gaps in knowledge, particularly in comparison with cytochrome P450 (CYP) enzymes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    阿尔茨海默病(AD)是一种神经退行性病理实体,其特征是tau和大分子Aβ沉积的异常存在导致神经元变性或死亡。除此之外,葡萄糖-6-磷酸脱氢酶(G6PD)在AD的发展过程中具有多方面的作用,其中它可以用作标记和目标。G6PD活性由于其对氧化应激的贡献而失调,神经炎症,和神经元死亡。在这种情况下,本综述生动地描述了有关AD进展与G6PD表达或活性变化之间关系的最新发现。在使用AD小鼠模型作为与该疾病相关的认知衰退和神经变性的代表性动物模型系统的多项研究中已经证明了所提出的基于G6PD的治疗剂的功效。通过新颖的创新纳米技术和药物管理技术中的微流体工具,为提高G6PD活性提供了创新的治疗见解。这些方法提供了超越血脑屏障的创新方法,靶向逐步特定的神经通路,并克服伴随AD的生化障碍。使用负载G6DP的不同纳米粒子靶向特定器官,例如,G6DP脂质体,增强BBB渗透和G6DP的脑分布。许多纳米粒子,用于不同的目的,在本文中进行了简要讨论。这种模拟芯片上器官BBB的方法提供了使用微流控芯片进行精确的疾病建模和药物测试,与传统技术相比,需要更低的样本量和更快的发现。简要地讨论了AD中微流体的其他贡献。然而,有一些限制伴随的微流体需要工作,以用于AD。这项研究旨在通过协同使用有前途的技术来弥合理解AD的差距;微流体和纳米技术,以促进未来的发展。
    Alzheimer\'s disease (AD) is a neurodegenerative pathologic entity characterized by the abnormal presence of tau and macromolecular Aβ deposition that leads to the degeneration or death of neurons. In addition to that, glucose-6-phosphate dehydrogenase (G6PD) has a multifaceted role in the process of AD development, where it can be used as both a marker and a target. G6PD activity is dysregulated due to its contribution to oxidative stress, neuroinflammation, and neuronal death. In this context, the current review presents a vivid depiction of recent findings on the relationship between AD progression and changes in the expression or activity of G6PD. The efficacy of the proposed G6PD-based therapeutics has been demonstrated in multiple studies using AD mouse models as representative animal model systems for cognitive decline and neurodegeneration associated with this disease. Innovative therapeutic insights are made for the boosting of G6PD activity via novel innovative nanotechnology and microfluidics tools in drug administration technology. Such approaches provide innovative methods of surpassing the blood-brain barrier, targeting step-by-step specific neural pathways, and overcoming biochemical disturbances that accompany AD. Using different nanoparticles loaded with G6DP to target specific organs, e.g., G6DP-loaded liposomes, enhances BBB penetration and brain distribution of G6DP. Many nanoparticles, which are used for different purposes, are briefly discussed in the paper. Such methods to mimic BBB on organs on-chip offer precise disease modeling and drug testing using microfluidic chips, requiring lower sample amounts and producing faster findings compared to conventional techniques. There are other contributions to microfluid in AD that are discussed briefly. However, there are some limitations accompanying microfluidics that need to be worked on to be used for AD. This study aims to bridge the gap in understanding AD with the synergistic use of promising technologies; microfluid and nanotechnology for future advancements.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:患有阿尔茨海默病(AD)的人通常需要许多药物;然而,这些药物使用推荐用于无AD患者的方案给药.尽管AD中血脑屏障(BBB)中P-糖蛋白(P-gp)的丰度和功能降低,这会影响大脑暴露的药物。导致散发性AD中P-gp丰度降低的基本机制仍然未知;然而,已知载脂蛋白E(apoE)基因与散发性AD的发生具有最强的遗传联系,和apoE亚型可以不同地改变BBB功能。这项研究的目的是使用人脑微血管内皮细胞(hCMEC/D3)模型评估apoE是否以同工型依赖性方式影响P-gp丰度和功能。
    方法:本研究评估了apoE亚型对hCMEC/D3细胞中P-gp丰度(通过蛋白质印迹)和功能(通过罗丹明123(R123)摄取)的影响。将细胞以2-10μg/mL暴露于重组apoE3和apoE424-72小时。hCMEC/D3细胞也暴露于来自表达人源化apoE同种型的星形胶质细胞的星形胶质细胞条件培养基(ACM)72小时。
    结果:相对于重组apoE3,重组apoE4没有改变hCMEC/D3细胞中的P-gp丰度,含有人apoE亚型的ACM也没有改变P-gp丰度。重组apoE同种型处理在hCMEC/D3细胞中的R123积累也没有改变,表明P-gp功能没有变化,尽管丰度和功能都被阳性对照SR12813(5µM)和PSC833(5µM)改变,分别。
    结论:在该模型中,不同的apoE同工型对P-gp丰度或功能没有直接影响,需要进一步的体内研究来解决散发性AD中P-gp丰度或功能是否以apoE同工型特异性方式降低。
    BACKGROUND: Individuals with Alzheimer\'s disease (AD) often require many medications; however, these medications are dosed using regimens recommended for individuals without AD. This is despite reduced abundance and function of P-glycoprotein (P-gp) at the blood-brain barrier (BBB) in AD, which can impact brain exposure of drugs. The fundamental mechanisms leading to reduced P-gp abundance in sporadic AD remain unknown; however, it is known that the apolipoprotein E (apoE) gene has the strongest genetic link to sporadic AD development, and apoE isoforms can differentially alter BBB function. The aim of this study was to assess if apoE affects P-gp abundance and function in an isoform-dependent manner using a human cerebral microvascular endothelial cell (hCMEC/D3) model.
    METHODS: This study assessed the impact of apoE isoforms on P-gp abundance (by western blot) and function (by rhodamine 123 (R123) uptake) in hCMEC/D3 cells. Cells were exposed to recombinant apoE3 and apoE4 at 2 - 10 µg/mL over 24 - 72 hours. hCMEC/D3 cells were also exposed for 72 hours to astrocyte-conditioned media (ACM) from astrocytes expressing humanised apoE isoforms.
    RESULTS: P-gp abundance in hCMEC/D3 cells was not altered by recombinant apoE4 relative to recombinant apoE3, nor did ACM containing human apoE isoforms alter P-gp abundance. R123 accumulation in hCMEC/D3 cells was also unchanged with recombinant apoE isoform treatments, suggesting no change to P-gp function, despite both abundance and function being altered by positive controls SR12813 (5 µM) and PSC 833 (5 µM), respectively.
    CONCLUSIONS: Different apoE isoforms have no direct influence on P-gp abundance or function within this model, and further in vivo studies would be required to address whether P-gp abundance or function are reduced in sporadic AD in an apoE isoform-specific manner.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    代谢综合征(例如,肥胖)的特征是胰岛素抵抗,慢性炎症,葡萄糖代谢受损,和血脂异常。最近,代谢综合征患者不仅经历了代谢问题,还经历了神经病理问题,包括认知障碍。多项研究报道了肥胖和糖尿病患者大脑中的血脑屏障(BBB)破坏和胰岛素抵抗。腺苷,嘌呤核苷,已知调节各种细胞反应(例如,神经炎症反应)通过与中枢神经系统(CNS)中的腺苷受体结合。腺苷有四种已知的受体:A1R,A2AR,A2BR,A3R这些受体在大脑的各种生理和病理过程中起着不同的作用,包括内皮细胞稳态,胰岛素敏感性,小胶质细胞激活,脂质代谢,免疫细胞浸润,和突触可塑性。这里,我们回顾了腺苷受体介导的信号在代谢失衡相关的神经病理学问题中的作用的最新发现.我们强调了腺苷信号传导在代谢综合征患者神经病理学问题治疗方案开发中的重要性。
    Metabolic syndromes (e.g., obesity) are characterized by insulin resistance, chronic inflammation, impaired glucose metabolism, and dyslipidemia. Recently, patients with metabolic syndromes have experienced not only metabolic problems but also neuropathological issues, including cognitive impairment. Several studies have reported blood-brain barrier (BBB) disruption and insulin resistance in the brain of patients with obesity and diabetes. Adenosine, a purine nucleoside, is known to regulate various cellular responses (e.g., the neuroinflammatory response) by binding with adenosine receptors in the central nervous system (CNS). Adenosine has four known receptors: A1R, A2AR, A2BR, and A3R. These receptors play distinct roles in various physiological and pathological processes in the brain, including endothelial cell homeostasis, insulin sensitivity, microglial activation, lipid metabolism, immune cell infiltration, and synaptic plasticity. Here, we review the recent findings on the role of adenosine receptor-mediated signaling in neuropathological issues related to metabolic imbalance. We highlight the importance of adenosine signaling in the development of therapeutic solutions for neuropathological issues in patients with metabolic syndromes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    缺血性脑卒中是一种死亡率和致残率较高的严重疾病。然而,临床上很少有神经保护药物用于缺血性卒中。造成这种失败的主要原因可能有两个:难以穿透血脑屏障(BBB)以及在血液循环中容易失活。Ferroptosis,脂质氧化相关的细胞死亡,在脑缺血再灌注损伤中起重要作用。我们利用RVG29,一种来自狂犬病病毒糖蛋白的肽,获得BBB靶向的脂质纳米粒(T-LNPs),以研究T-LNPs是否改善了Ferrostatin-1(Fer1,铁凋亡的抑制剂)对脑缺血损伤的神经保护作用。T-LNP在体外BBB模型中暴露于氧/葡萄糖剥夺后显着增加了BBB的渗透,并在脑缺血鼠模型中给药后6小时增强了脑组织中的荧光分布。此外,T-LNPs包裹的Fer1(T-LNPs-Fer1)通过维持脑缺血后神经元细胞中NADPH氧化酶4(NOX4)和谷胱甘肽过氧化物酶4(GPX4)信号的稳态,显着增强了Fer1对铁凋亡的抑制作用。T-LNPs-Fer1显著抑制神经元的氧化应激[血红素加氧酶-1表达和丙二醛(脂质ROS反应的产物)],减轻缺血诱导的神经元细胞死亡,与未封装的单独Fer1相比。此外,在45分钟缺血/24小时再灌注后,与Fer1处理的脑缺血小鼠相比,T-LNP-Fer1显着减少了脑梗死并改善了行为功能。这些发现表明,T-LNPs有助于Fer1穿透BBB并改善Fer1对实验性中风中脑缺血损伤的神经保护作用,为临床治疗缺血性卒中药物的开发提供可行的转化策略。
    Cerebral ischemic stroke is a serious disease with high mortality and disability rates. However, few neuroprotective drugs have been used for ischemic stroke in the clinic. Two main reasons may be responsible for this failure: difficulty in penetrating the blood-brain barrier (BBB) and easily inactivated in the blood circulation. Ferroptosis, a lipid oxidation-related cell death, plays significant roles in cerebral ischemia-reperfusion injury. We utilized RVG29, a peptide derived from Rabies virus glycoprotein, to obtain BBB-targeted lipid nanoparticles (T-LNPs) in order to investigate whether T-LNPs improved the neuroprotective effects of Ferrostatin-1 (Fer1, an inhibitor of ferroptosis) against cerebral ischemic damage. T-LNPs significantly increased BBB penetration following oxygen/glucose deprivation exposure in an in vitro BBB model and enhanced the fluorescence distribution in brain tissues at 6 h post-administration in a cerebral ischemic murine model. Moreover, T-LNPs encapsulated Fer1 (T-LNPs-Fer1) significantly enhanced the inhibitory effects of Fer1 on ferroptosis by maintaining the homeostasis of NADPH oxidase 4 (NOX4) and glutathione peroxidase 4 (GPX4) signals in neuronal cells after cerebral ischemia. T-LNPs-Fer1 significantly suppressed oxidative stress [heme oxygenase-1 expression and malondialdehyde (the product of lipid ROS reaction)] in neurons and alleviated ischemia-induced neuronal cell death, compared to Fer1 alone without encapsulation. Furthermore, T-LNPs-Fer1 significantly reduced cerebral infarction and improved behavior functions compared to Fer1-treated cerebral ischemic mice after 45-min ischemia/24-h reperfusion. These findings showed that the T-LNPs helped Fer1 penetrate the BBB and improved the neuroprotection of Fer1 against cerebral ischemic damage in experimental stroke, providing a feasible translational strategy for the development of clinical drugs for the treatment of ischemic stroke.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    弥漫性内在脑桥神经胶质瘤(DIPG)是一种侵袭性脑肿瘤,发生在脑干脑桥中,占所有脑干神经胶质瘤的80%以上。诊断时的中位年龄为6-7岁,诊断后2年总生存率低于10%,5年后总生存率低于1%。DIPG手术无法进入,放射治疗只能提供短暂的益处,持续的局部肿瘤浸润导致死亡。DIPGs现在是儿童脑瘤死亡的主要原因,每个人的社会癌症负担超过67岁的生命损失(YLL),肺癌和乳腺癌分别为14和16YLL。已经对患有DIPG的儿童进行了95多项临床药物试验,所有这些都未能改善生存。迄今为止,由于我们无法确定针对该疾病的靶向药物并无法通过完整的血脑屏障(BBB)递送这些药物,因此尚无单一或联合化疗策略成功。因此,DIPG越来越关注免疫治疗研究,随着对嵌合抗原受体T(CAR-T)细胞等治疗方法的探索,免疫检查点封锁,癌症疫苗,和自体细胞转移治疗。这里,我们回顾了在确定影响DIPG免疫治疗发展的遗传因素方面的最新进展.此外,我们探索新兴技术,如磁共振引导聚焦超声(MRgFUS)在潜在的组合方法来治疗DIPG。
    Diffuse intrinsic pontine glioma (DIPG) is an aggressive brain tumour that occurs in the pons of the brainstem and accounts for over 80% of all brainstem gliomas. The median age at diagnosis is 6-7 years old, with less than 10% overall survival 2 years after diagnosis and less than 1% after 5 years. DIPGs are surgically inaccessible, and radiation therapy provides only transient benefit, with death ensuing from relentless local tumour infiltration. DIPGs are now the leading cause of brain tumour deaths in children, with a societal cancer burden in years of life lost (YLL) of more than 67 per individual, versus approximately 14 and 16 YLL for lung and breast cancer respectively. More than 95 clinical drug trials have been conducted on children with DIPGs, and all have failed to improve survival. No single or combination chemotherapeutic strategy has been successful to date because of our inability to identify targeted drugs for this disease and to deliver these drugs across an intact blood-brain barrier (BBB). Accordingly, there has been an increased focus on immunotherapy research in DIPG, with explorations into treatments such as chimeric antigen receptor T (CAR-T) cells, immune checkpoint blockades, cancer vaccines, and autologous cell transfer therapy. Here, we review the most recent advances in identifying genetic factors influencing the development of immunotherapy for DIPG. Additionally, we explore emerging technologies such as Magnetic Resonance-guided Focused Ultrasound (MRgFUS) in potential combinatorial approaches to treat DIPG.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    P-糖蛋白(P-gp)和乳腺癌耐药蛋白(BCRP)多药耐药(MDR)转运蛋白位于血脑屏障(BBB)的腔表面。它们赋予胎儿大脑对可能在外周血中循环的有害化合物的保护作用。胎儿在低氧水平下发育;然而,一些产科病变,如先兆子痫,胎盘植入/前置可能导致更大的胎儿缺氧状态。我们研究了缺氧如何影响妊娠早期和中期来源的人胎儿脑内皮细胞(hfBECs)中的MDR转运蛋白。缺氧降低了孕早期hfBECs中的BCRP蛋白和活性。相比之下,在妊娠中期的hfBECs中,缺氧后P-gp和BCRP活性增加。结果表明,在妊娠早期,缺氧引起的胎儿脑保护作用降低,但在妊娠中期,转运蛋白介导的BBB保护作用可能增加。这将改变P-gp和BCRP的各种关键生理和药理学底物在发育中的胎儿脑中的积累,并可能导致通常与子宫内缺氧相关的神经发育障碍的发病机理。
    P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) multidrug resistance (MDR) transporters are localized at the luminal surface of the blood-brain barrier (BBB). They confer fetal brain protection against harmful compounds that may be circulating in the peripheral blood. The fetus develops in low oxygen levels; however, some obstetric pathologies such as pre-eclampsia, placenta accreta/previa may result in even greater fetal hypoxic states. We investigated how hypoxia impacts MDR transporters in human fetal brain endothelial cells (hfBECs) derived from early and mid-stages of pregnancy. Hypoxia decreased BCRP protein and activity in hfBECs derived in early pregnancy. In contrast, in hfBECs derived in mid-pregnancy there was an increase in P-gp and BCRP activity following hypoxia. Results suggest a hypoxia-induced reduction in fetal brain protection in early pregnancy, but a potential increase in transporter-mediated protection at the BBB during mid-gestation. This would modify accumulation of various key physiological and pharmacological substrates of P-gp and BCRP in the developing fetal brain and potentially contribute to the pathogenesis of neurodevelopmental disorders commonly associated with in utero hypoxia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    众所周知,药物治疗在脑缺血的治疗和预防中起着关键作用。然而,现有药物,包括许多天然产品,应用于脑缺血治疗时遇到各种挑战。这些挑战包括由于低血脑屏障(BBB)渗透性导致的大脑吸收不良,有限的水溶性,生物利用度不足,稳定性差,和快速的新陈代谢。为了解决这些问题,研究人员已经转向前药策略,旨在减轻或消除母体药物分子的不利性质。体内代谢或酶促反应将前药转化为活性母体药物,从而增加BBB通透性,提高生物利用度和稳定性,减少对正常组织的毒性,最终旨在提高治疗的有效性和安全性。这篇全面的综述探讨了多种有效的前药策略,提供了在过去二十年中开发的代表性前药的详细描述。它强调了前药方法改善目前可用的脑缺血药物的治疗结果的潜力。这篇综述的发表有助于丰富当前关于治疗和预防脑缺血的前药策略的研究进展。此外,它旨在为该领域的药物化学家提供有价值的见解,为脑缺血药物的开发提供指导,并为患者提供更安全、更有效的药物治疗选择。
    It is well-known that pharmacotherapy plays a pivotal role in the treatment and prevention of cerebral ischemia. Nevertheless, existing drugs, including numerous natural products, encounter various challenges when applied in cerebral ischemia treatment. These challenges comprise poor brain absorption due to low blood-brain barrier (BBB) permeability, limited water solubility, inadequate bioavailability, poor stability, and rapid metabolism. To address these issues, researchers have turned to prodrug strategies, aiming to mitigate or eliminate the adverse properties of parent drug molecules. In vivo metabolism or enzymatic reactions convert prodrugs into active parent drugs, thereby augmenting BBB permeability, improving bioavailability and stability, and reducing toxicity to normal tissues, ultimately aiming to enhance treatment efficacy and safety. This comprehensive review delves into multiple effective prodrug strategies, providing a detailed description of representative prodrugs developed over the past two decades. It underscores the potential of prodrug approaches to improve the therapeutic outcomes of currently available drugs for cerebral ischemia. The publication of this review serves to enrich current research progress on prodrug strategies for the treatment and prevention of cerebral ischemia. Furthermore, it seeks to offer valuable insights for pharmaceutical chemists in this field, offer guidance for the development of drugs for cerebral ischemia, and provide patients with safer and more effective drug treatment options.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号