In vivo efficacy

体内功效
  • 文章类型: Journal Article
    有机金属化合物,包括钌配合物,已经被广泛开发为抗癌化疗药物,但作为潜在的抗寄生虫药物也引起了极大的兴趣。最近合成了由与不同抗微生物剂络合的有机金属钌部分组成的杂合药物。其中一种化合物,与磺胺多辛(SDX)共轭的三硫醇-二钌络合物(RU),抑制人包皮成纤维细胞(HFF)单层中生长的弓形虫速殖子的增殖,IC50<150nM,而单独或作为等摩尔混合物应用的SDX和未修饰的RU复合物的效力低得多。此外,SDX与RU的缀合导致HFF细胞毒性降低。RU-SDX在0.1至0.5μM的浓度范围内不会损害小鼠脾细胞的体外增殖,但在2μM时具有影响,并在20μM诱导斑马鱼胚胎毒性,但不是2或0.2μM。RU-SDX具有抗寄生虫作用,但不具有杀寄生虫作用,并在治疗早期诱导速殖子线粒体基质的瞬时超微结构变化。虽然其他靶向线粒体的化合物,如解偶联剂FCCP和CCCP以及与腺嘌呤缀合的另一种三硫代钌复合物影响线粒体膜电位,RU-SDX未检测到这种效应.评估RU-SDX在由非妊娠近交CD1小鼠组成的鼠弓形虫卵囊感染模型中的体内功效,显示对脑寄生虫负荷没有影响,但减少了眼睛和心脏组织中的寄生虫负荷。
    Organometallic compounds, including Ruthenium complexes, have been widely developed as anti-cancer chemotherapeutics, but have also attracted much interest as potential anti-parasitic drugs. Recently hybrid drugs composed of organometallic Ruthenium moieties that were complexed to different antimicrobial agents were synthesized. One of these compounds, a trithiolato-diRuthenium complex (RU) conjugated to sulfadoxine (SDX), inhibited proliferation of Toxoplasma gondii tachyzoites grown in human foreskin fibroblast (HFF) monolayers with an IC50 < 150 nM, while SDX and the non-modified RU complex applied either individually or as an equimolar mixture were much less potent. In addition, conjugation of SDX to RU lead to decreased HFF cytotoxicity. RU-SDX did not impair the in vitro proliferation of murine splenocytes at concentrations ranging from 0.1 to 0.5 μM but had an impact at 2 μM, and induced zebrafish embryotoxicity at 20 μM, but not at 2 or 0.2 μM. RU-SDX acted parasitostatic but not parasiticidal, and induced transient ultrastructural changes in the mitochondrial matrix of tachyzoites early during treatment. While other compounds that target the mitochondrion such as the uncouplers FCCP and CCCP and another trithiolato-Ruthenium complex conjugated to adenine affected the mitochondrial membrane potential, no such effect was detected for RU-SDX. Evaluation of the in vivo efficacy of RU-SDX in a murine T. gondii oocyst infection model comprised of non-pregnant outbred CD1 mice showed no effects on the cerebral parasite burden, but reduced parasite load in the eyes and in heart tissue.
    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
    弓形虫病是一种全球流行的人畜共患疾病,具有重要的临床意义。包括神经弓形虫病,艾滋病患者脑部病变的主要原因。目前的药物治疗弓形虫病面临临床局限性,迫切需要开发新的疗法。天然来源产生了多种生物活性化合物,作为临床使用的衍生物的基础。海洋细菌衍生的天然产物的探索导致了marinoquinolines,其特征是吡咯并喹啉核心,并在体外和体内表现出抗疟原虫活性。这项研究调查了六种海藻喹啉衍生物的体外抗弓形虫潜力。此外,它进行吸收,分布,新陈代谢,排泄,和毒性(ADMET)预测,并评价一种选定化合物的体内功效。这些化合物显示出1.31至3.78µM的半最大有效浓度(EC50)值和4.16至30.51µM的半最大细胞毒性浓度(CC50)值。导致选择性指数(SI)从3.18到20.85。MQ-1表现出最高的体外SI,以12.5mg/kg/天的剂量连续八天口服时,RH感染的瑞士小鼠腹膜中的速殖子数量显着减少。此外,MQ-1以25mg/kg/天的剂量连续10天口服给药时,可显着降低慢性ME49感染的C57BL/6小鼠的脑寄生虫负担。这些发现强调了有希望的反T。marinoquinolines的刚地活性及其作为该疾病的新型治疗剂的潜力。
    Toxoplasmosis is a globally prevalent zoonotic disease with significant clinical implications, including neurotoxoplasmosis, a leading cause of cerebral lesions in AIDS patients. The current pharmacological treatments for toxoplasmosis face clinical limitations, necessitating the urgent development of new therapeutics. Natural sources have yielded diverse bioactive compounds, serving as the foundation for clinically used derivatives. The exploration of marine bacteria-derived natural products has led to marinoquinolines, which feature a pyrroloquinoline core and demonstrate in vitro and in vivo anti-Plasmodium activity. This study investigates the in vitro anti-Toxoplasma gondii potential of six marinoquinoline derivatives. Additionally, it conducts absorption, distribution, metabolism, excretion, and toxicity (ADMET) predictions, and evaluates the in vivo efficacy of one selected compound. The compounds displayed half-maximal effective concentration (EC50) values between 1.31 and 3.78 µM and half-maximal cytotoxic concentration (CC50) values ranging from 4.16 to 30.51 µM, resulting in selectivity indices (SI) from 3.18 to 20.85. MQ-1 exhibiting the highest in vitro SI, significantly reduced tachyzoite numbers in the peritoneum of RH-infected Swiss mice when it was orally administered at 12.5 mg/kg/day for eight consecutive days. Also, MQ-1 significantly reduced the cerebral parasite burden in chronically ME49 infected C57BL/6 mice when it was orally administered at 25 mg/kg/day for 10 consecutive days. These findings underscore the promising anti-T. gondii activity of marinoquinolines and their potential as novel therapeutic agents against this disease.
    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
    类风湿性关节炎是一种全身性自身免疫性炎性疾病,影响全球数百万人。有多种改善疾病的抗风湿药物可用;然而,许多患者对任何治疗都没有反应。由于解整合素和金属蛋白酶10在从细胞表面释放多种促炎和抗炎因子中的作用,因此已被认为是RA的潜在新靶标。在本研究中,我们测定了一类新型非锌结合抑制剂的化合物CID3117694的药代动力学参数和体内疗效.口服生物利用度在10mg/kg剂量后在血液和滑液中得到证实。为了测试功效,我们建立了胶原诱导的小鼠关节炎模型。CID3117694以10、30和50mg/kg/天口服给药28天。CID3117694能够剂量依赖性地改善疾病评分,减少血液中的RA标记,减少炎症的迹象,增生,血管nus形成,与未治疗的对照相比,受影响的关节中的软骨侵蚀。此外,用CID3117694治疗的小鼠没有表现出任何临床症状,表明低毒性。这项研究的结果表明,抑制ADAM10exosite可以是RA的可行治疗方法。
    Rheumatoid arthritis is a systemic autoimmune inflammatory disease that affects millions of people worldwide. There are multiple disease-modifying anti-rheumatic drugs available; however, many patients do not respond to any treatment. A disintegrin and metalloproteinase 10 has been suggested as a potential new target for RA due to its role in the release of multiple pro- and anti-inflammatory factors from cell surfaces. In the present study, we determined the pharmacokinetic parameters and in vivo efficacy of a compound CID3117694 from a novel class of non-zinc-binding inhibitors. Oral bioavailability was demonstrated in the blood and synovial fluid after a 10 mg/kg dose. To test efficacy, we established the collagen-induced arthritis model in mice. CID3117694 was administered orally at 10, 30, and 50 mg/kg/day for 28 days. CID3117694 was able to dose-dependently improve the disease score, decrease RA markers in the blood, and decrease signs of inflammation, hyperplasia, pannus formation, and cartilage erosion in the affected joints compared to the untreated control. Additionally, mice treated with CID 3117694 did not exhibit any clinical signs of distress, suggesting low toxicity. The results of this study suggest that the inhibition of ADAM10 exosite can be a viable therapeutic approach to RA.
    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
    在这项研究中,含有水溶性差的药物的无定形固体分散体,比沙科迪尔,通过热熔挤出制备以增强其治疗效果。首先,药物与聚合物之间的混溶性和相互作用作为预配制策略进行了研究,使用各种分析方法来获得选择合适聚合物的信息.基于Hansen溶解度参数的计算和单一玻璃化转变温度(Tg)的确定,bisacodyl和所有研究的聚合物之间的混溶性被证实。此外,基于动态蒸气吸附(DVS)的综合结果鉴定了药物-聚合物分子相互作用,傅里叶变换红外光谱(FT-IR),拉曼光谱,并比较了Tg的预测值和实验值。特别是,基于羟丙基甲基纤维素(HPMC)的固体分散体,在DVS实验后,Tg的计算值与实验值之间存在较大偏差,并且具有出色的物理稳定性,由于基于非汇溶出测试的结果对沉淀具有优异的抑制作用,因此被选择为最合适的溶解的比沙可啶制剂。此外,研究表明,与含有未溶解的生比沙可啶的制剂相比,含有1:4比例(w/w)的HPMC-比沙可啶的肠溶片在便秘诱导的兔子中的体内治疗性泻药功效显着提高。因此,得出的结论是,制定前的战略,使用几种分析和方法,在这项研究中成功地应用于研究药物-聚合物系统的混溶性和相互作用,因此导致使用热熔挤出方法制造具有有利的体外和体内性能的有利的固体分散体。
    In this study, an amorphous solid dispersion containing the poorly water-soluble drug, bisacodyl, was prepared by hot-melt extrusion to enhance its therapeutic efficacy. First, the miscibility and interaction between the drug and polymer were investigated as pre-formulation strategies using various analytical approaches to obtain information for selecting a suitable polymer. Based on the calculation of the Hansen solubility parameter and the identification of the single glass transition temperature (Tg), the miscibility between bisacodyl and all the investigated polymers was confirmed. Additionally, the drug-polymer molecular interaction was identified based on the comprehensive results of dynamic vapor sorption (DVS), Fourier transform infrared spectroscopy (FT-IR), Raman spectroscopy, and a comparison of the predicted and experimental values of Tg. In particular, the hydroxypropyl methylcellulose (HPMC)-based solid dispersions, which exhibited large deviation between the calculated and experimental values of Tg and superior physical stability after DVS experiments, were selected as the most appropriate solubilized bisacodyl formulations due to the excellent inhibitory effects on precipitation based on the results of the non-sink dissolution test. Furthermore, it was shown that the enteric-coated tablets containing HPMC-bisacodyl at a 1:4 ratio (w/w) had significantly improved in vivo therapeutic laxative efficacy compared to preparations containing un-solubilized raw bisacodyl in constipation-induced rabbits. Therefore, it was concluded that the pre-formulation strategy, using several analyses and approaches, was successfully applied in this study to investigate the miscibility and interaction of drug-polymer systems, hence resulting in the manufacture of favorable solid dispersions with favorable in vitro and in vivo performances using hot-melt extrusion processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基孔肯雅病毒是一种重新出现的虫媒病毒,近几十年来引起了流行病的爆发。在急性感染期间,高病毒载量和严重免疫反应的老年患者可能会发展为慢性关节炎和严重的关节痛。目前,没有抗病毒药物可用。以前的研究表明,黄酮衍生物,8-bromobaicalein,是靶向黄病毒聚合酶的基于细胞的系统中潜在的登革热和寨卡病毒复制抑制剂。在这里,我们表征了8-溴黄芩素在Vero细胞中以0.49±0.11µM的EC50抑制基孔肯雅病毒复制。使用分子结合和片段分子轨道计算预测病毒nsP1甲基转移酶的分子靶标。此外,口服250mg/kg每日两次治疗可减轻基孔肯雅诱导的肌肉骨骼炎症,并降低健康成年小鼠的病毒载量。药代动力学分析表明,250mg/kg给药使化合物水平维持在EC99.9以上12小时。8-溴黄芩素应该是进一步开发为泛虫媒病毒药物的潜在候选者。
    Chikungunya virus is a re-emerging arbovirus that has caused epidemic outbreaks in recent decades. Patients in older age groups with high viral load and severe immunologic response during acute infection are likely to develop chronic arthritis and severe joint pain. Currently, no antiviral drug is available. Previous studies suggested that a flavone derivative, 8-bromobaicalein, was a potential dengue and Zika replication inhibitor in a cell-based system targeting flaviviral polymerase. Here we characterized that 8-bromobaicalein inhibited chikungunya virus replication with EC50 of 0.49 ± 0.11 µM in Vero cells. The molecular target predicted at viral nsP1 methyltransferase using molecular binding and fragment molecular orbital calculation. Additionally, oral administration of 250 mg/kg twice daily treatment alleviated chikungunya-induced musculoskeletal inflammation and reduced viral load in healthy adult mice. Pharmacokinetic analysis indicated that the 250 mg/kg administration maintained the compound level above EC99.9 for 12 h. Therefore, 8-bromobaicalein should be a potential candidate for further development as a pan-arboviral drug.
    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
    烟曲霉的唑耐药性是全世界关注的问题,需要新的抗真菌药物来克服这个问题。他汀,3-羟基-3-甲基戊二酰辅酶A(HMG-CoA)还原酶抑制剂,据报道可以抑制烟曲霉的生长,但对其对烟曲霉的体内抗真菌作用知之甚少。在这项研究中,我们用家蚕模型评估了匹伐他汀(PIT)联合伊曲康唑(ITC)对唑类敏感和唑类耐药菌株的体内疗效。在唑敏感和唑耐药菌株模型中,与未治疗组相比,联合治疗(PIT和ITC)组的生存期延长。此外,当使用唑敏感菌株时,与单独使用ITC相比,联合治疗可获得更高的生存率.蚕的组织病理学分析显示,在唑敏感和唑耐药菌株模型中,菌丝量减少。通过qPCR在唑敏感菌株模型中定量评估真菌DNA,阐明了与未治疗组和单独ITC组相比,联合治疗组的真菌负荷降低。这些结果表明,当在体内与ITC组合时,PIT的功效增强。与大多数他汀类药物相反,PIT与人的唑类药物几乎没有药物相互作用,可以安全地与ITC一起使用。这种联合疗法可能是未来临床环境中有效治疗的有希望的选择。重要性烟曲霉分离株中的唑耐药性最近越来越被认为是治疗失败的原因。和替代抗真菌疗法需要克服这个问题。我们的研究表明,通过几种方法,包括评估存活率,使用家蚕模型,PIT联合ITC对烟曲霉的体内功效。组织病理学分析,和真菌负荷的评估。与大多数他汀类药物相反,PIT可以安全地使用唑类药物,因为药物-药物相互作用较少,因此,这项研究将有助于我们验证如何在未来的临床环境中使用该药物。
    Azole resistance in Aspergillus fumigatus is a worldwide concern and new antifungal drugs are required to overcome this problem. Statin, a 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor, has been reported to suppress the growth of A. fumigatus, but little is known about its in vivo antifungal effect against A. fumigatus. In this study, we evaluated the in vivo efficacy of pitavastatin (PIT) combined with itraconazole (ITC) against azole-susceptible and azole-resistant strains with silkworm models. Prolongation of survival was confirmed in the combination-therapy (PIT and ITC) group compared to the no-treatment group in both azole-susceptible and azole-resistant strain models. Furthermore, when the azole-susceptible strain was used, the combination-therapy resulted in a higher survival rate than with ITC alone. Histopathological analysis of the silkworms revealed a reduction of the hyphal amount in both azole-susceptible and azole-resistant strain models. Quantitative evaluation of fungal DNA by qPCR in azole-susceptible strain models clarified the reduction of fungal burden in the combination-therapy group compared with the no-treatment group and ITC-alone group. These results indicate that the efficacy of PIT was enhanced when combined with ITC in vivo. As opposed to most statins, PIT has little drug-drug interaction with azoles in humans and can be used safely with ITC. This combination therapy may be a promising option as an effective treatment in clinical settings in the future. IMPORTANCE Azole resistance among A. fumigatus isolates has recently been increasingly recognized as a cause of treatment failure, and alternative antifungal therapies are required to overcome this problem. Our study shows the in vivo efficacy of PIT combined with ITC against A. fumigatus using silkworm models by several methods including evaluation of survival rates, histopathological analysis, and assessment of fungal burden. Contrary to most statins, PIT can be safely administered with azoles because of less drug-drug interactions, so this study should help us to verify how to make use of the drug in clinical settings in the future.
    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
    金黄色葡萄球菌引起广泛的感染,它是导致抗生素耐药性相关死亡的主要病原体之一,金黄色葡萄球菌的迅速传播促使人们发现新的抗生素。新候选药物的体内功效的评估通常使用动物模型进行。最近,斑马鱼(Daniorerio)胚胎在早期药物发现中变得越来越有吸引力。在这里,我们建立了金黄色葡萄球菌感染斑马鱼胚胎模型,用于评价新型潜在抗菌药物的体内疗效.通过显微注射表达mCherry的金黄色葡萄球菌Newman,然后通过显微注射到不同的注射部位以及通过水性暴露用参考抗生素治疗来诱导局部感染,以研究给药途径对功效的影响。我们成功地使用开发的模型来评估天然产物索兰霉素A的体内活性,由于血浆快速降解,普通小鼠模型不成功。总之,我们提出了一个新的筛选平台,用于评估抗生素发现阶段的体内活性。此外,这项工作为根据测试药物的理化性质选择合适的给药途径提供了考虑。
    Staphylococcus aureus causes a wide range of infections, and it is one of the leading pathogens responsible for deaths associated with antimicrobial resistance, the rapid spread of which among S. aureus urges the discovery of new antibiotics. The evaluation of in vivo efficacy of novel drug candidates is usually performed using animal models. Recently, zebrafish (Danio rerio) embryos have become increasingly attractive in early drug discovery. Herein, we established a zebrafish embryo model of S. aureus infection for evaluation of in vivo efficacy of novel potential antimicrobials. A local infection was induced by microinjecting mCherry-expressing S. aureus Newman followed by treatment with reference antibiotics via microinjection into different injection sites as well as via waterborne exposure to study the impact of the administration route on efficacy. We successfully used the developed model to evaluate the in vivo activity of the natural product sorangicin A, for which common mouse models were not successful due to fast degradation in plasma. In conclusion, we present a novel screening platform for assessing in vivo activity at the antibiotic discovery stage. Furthermore, this work provides consideration for the choice of an appropriate administration route based on the physicochemical properties of tested drugs.
    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
    在保留健康细胞和组织的同时将化学疗法选择性递送到肿瘤部位是癌症治疗的有吸引力的方法。诸如肽的载体可以促进选择性肿瘤靶向和有效载荷递送。将对癌细胞中过表达的细胞表面受体具有特异性亲和力的肽缀合至化学疗法以提供显示被癌细胞选择性摄取的肽-药物缀合物(PDC)。使用名为18-4的10-mer线性肽(WxEAAYQrFL)靶向并结合乳腺癌细胞,我们设计了一种肽18-4-多柔比星(Dox)缀合物,对三阴性乳腺癌(TNBC)MDA-MB-231细胞具有高特异性毒性,对正常乳腺MCF10A上皮细胞具有30倍的毒性。这里,我们阐明了这种有效和肿瘤选择性肽18-4-Dox缀合物在携带原位MDA-MB-231肿瘤的小鼠中的体内活性。与用等效Dox剂量的游离Dox处理的小鼠相比,用四次每周注射缀合物处理的小鼠显示出显著更低的肿瘤体积。小鼠组织的免疫组织化学(IHC)分析显示,用低剂量的PDC(2.5mg/kg的Dox当量)处理降低了增殖标志物(PCNA和Ki-67)的表达并增加了细胞凋亡(通过增加的caspase-3表达证明)。在相同剂量的游离Dox(2.5mg/kg)下,这些标志物的表达与盐水处理相似。因此,与Dox处理的小鼠相比,缀合物处理的小鼠的肿瘤中积累的Dox明显更多(7倍),而在肝脏中观察到较低水平的Dox,心,和肽-Dox缀合物处理的小鼠的肺(比Dox处理的小鼠少3倍)。角蛋白1(K1)的IHC分析,肽18-4的受体揭示了K1在肿瘤中的上调和在正常乳腺脂肪垫和小鼠肝脏组织中的低水平,提示TNBC优先摄取PDC是K1受体介导的。一起来看,我们的数据支持使用PDC方法选择性地将化疗方案递送至TNBC以抑制肿瘤生长.
    Selective delivery of chemotherapy to the tumor site while sparing healthy cells and tissues is an attractive approach for cancer treatment. Carriers such as peptides can facilitate selective tumor targeting and payload delivery. Peptides with specific affinity for the overexpressed cell-surface receptors in cancer cells are conjugated to chemotherapy to afford peptide-drug conjugates (PDCs) that show selective uptake by cancer cells. Using a 10-mer linear peptide (WxEAAYQrFL) called 18-4 that targets and binds breast cancer cells, we designed a peptide 18-4-doxorubicin (Dox) conjugate with high specific toxicity toward triple-negative breast cancer (TNBC) MDA-MB-231 cells and 30-fold lower toxicity to normal breast MCF10A epithelial cells. Here, we elucidate the in vivo activity of this potent and tumor-selective peptide 18-4-Dox conjugate in mice bearing orthotopic MDA-MB-231 tumors. Mice treated with four weekly injections of the conjugate showed significantly lower tumor volumes compared to mice treated with free Dox at an equivalent Dox dose. Immunohistochemical (IHC) analysis of mice tissues revealed that treatment with a low dose of PDC (2.5 mg/kg of Dox equiv) reduced the expression of proliferation markers (PCNA and Ki-67) and increased apoptosis (evidenced by increased caspase-3 expression). At the same dose of free Dox (2.5 mg/kg), the expression of these markers was similar to that of saline treatment. Accordingly, significantly more Dox accumulated in tumors of conjugate-treated mice (7-fold) compared to the Dox-treated mice, while lower levels of Dox were observed in the liver, heart, and lungs of peptide-Dox conjugate-treated mice (up to 3-fold less) than Dox-treated mice. The IHC analysis of keratin 1 (K1), the receptor for peptide 18-4, revealed K1 upregulation in tumors and low levels in normal mammary fat pad and liver tissues from mice, suggesting preferential uptake of PDCs by TNBC to be K1 receptor-mediated. Taken together, our data support the use of a PDC approach to deliver chemotherapy selectively to the TNBC to inhibit tumor growth.
    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
    青蒿素(ART)是关键的抗疟疾药,尽管它们用于联合治疗,抗ART的恶性疟原虫正在全球蔓延。为了对抗ART抵抗,我们设计了阿替佐米(ATZs),通过非不稳定酰胺键将ART与蛋白酶体抑制剂(PI)连接的分子,并劫持寄生虫自身的泛素-蛋白酶体系统,以原位创建新型抗疟疾药。在ART部分激活后,ATZ共价附着并破坏多种寄生虫蛋白,标记它们的蛋白酶体退化。当受损的蛋白质进入蛋白酶体时,它们附着的PI抑制蛋白酶功能,增强ART的杀寄生虫作用并克服ART抗性。PI部分与蛋白酶体活性位点的结合通过延伸的连接肽的远端相互作用而增强。提供一种机制来克服PI阻力。除了每个组件之外,ATZ还有一个额外的行动模式,从而克服了对这两个组件的阻力,同时避免当单个药物具有不同的药代动力学特征时看到的短暂单一疗法。
    Artemisinins (ART) are critical anti-malarials and despite their use in combination therapy, ART-resistant Plasmodium falciparum is spreading globally. To counter ART resistance, we designed artezomibs (ATZs), molecules that link an ART with a proteasome inhibitor (PI) via a non-labile amide bond and hijack parasite\'s own ubiquitin-proteasome system to create novel anti-malarials in situ. Upon activation of the ART moiety, ATZs covalently attach to and damage multiple parasite proteins, marking them for proteasomal degradation. When damaged proteins enter the proteasome, their attached PIs inhibit protease function, potentiating the parasiticidal action of ART and overcoming ART resistance. Binding of the PI moiety to the proteasome active site is enhanced by distal interactions of the extended attached peptides, providing a mechanism to overcome PI resistance. ATZs have an extra mode of action beyond that of each component, thereby overcoming resistance to both components, while avoiding transient monotherapy seen when individual agents have disparate pharmacokinetic profiles.
    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
    WD重复结构域5(WDR5)是许多多蛋白复合物的核心支架组件,这些复合物在细胞核中执行各种关键的染色质中心过程。WDR5是混合谱系白血病MLL/SET复合物的组成部分,并将MYC定位于肿瘤关键靶基因的染色质。作为这些复合物的一部分,WDR5在维持多种人类癌症的肿瘤发生中起作用,这些癌症通常与不良预后有关。因此,WDR5已被认为是治疗实体肿瘤和血液肿瘤的有吸引力的治疗靶标。以前,WDR5-相互作用(WIN)位点的小分子抑制剂和WDR5降解剂在癌细胞系中显示出强大的体外细胞功效,并确立了WDR5的治疗潜力.然而,在动物疾病模型中,通过口服给药,这些药物在药理学相关剂量下未显示出显著的体内功效.我们已经发现WDR5WIN位点抑制剂,其通过基于结构的设计以双环杂芳基P7单元为特征,并且解决我们先前系列的小分子抑制剂的局限性。重要的是,我们的先导化合物表现出增强的目标效能,优异的口服药代动力学(PK)概况,以及通过口服给药在小鼠MV4:11皮下异种移植模型中的有效剂量依赖性体内功效。此外,这些体内探针在啮齿类动物的重复高剂量方案下显示出优异的耐受性,证明了WDR5WIN位点抑制机制的安全性.总的来说,我们的结果为WDR5WIN位点抑制剂用作潜在的抗癌治疗药物提供了有力支持.
    WD repeat domain 5 (WDR5) is a core scaffolding component of many multiprotein complexes that perform a variety of critical chromatin-centric processes in the nucleus. WDR5 is a component of the mixed lineage leukemia MLL/SET complex and localizes MYC to chromatin at tumor-critical target genes. As a part of these complexes, WDR5 plays a role in sustaining oncogenesis in a variety of human cancers that are often associated with poor prognoses. Thus, WDR5 has been recognized as an attractive therapeutic target for treating both solid and hematological tumors. Previously, small-molecule inhibitors of the WDR5-interaction (WIN) site and WDR5 degraders have demonstrated robust in vitro cellular efficacy in cancer cell lines and established the therapeutic potential of WDR5. However, these agents have not demonstrated significant in vivo efficacy at pharmacologically relevant doses by oral administration in animal disease models. We have discovered WDR5 WIN-site inhibitors that feature bicyclic heteroaryl P7 units through structure-based design and address the limitations of our previous series of small-molecule inhibitors. Importantly, our lead compounds exhibit enhanced on-target potency, excellent oral pharmacokinetic (PK) profiles, and potent dose-dependent in vivo efficacy in a mouse MV4:11 subcutaneous xenograft model by oral dosing. Furthermore, these in vivo probes show excellent tolerability under a repeated high-dose regimen in rodents to demonstrate the safety of the WDR5 WIN-site inhibition mechanism. Collectively, our results provide strong support for WDR5 WIN-site inhibitors to be utilized as potential anticancer therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号