In vivo efficacy

体内功效
  • 文章类型: Journal Article
    尽管三代表皮生长因子受体(EGFR)-TK抑制剂已被批准用于治疗非小细胞肺癌(NSCLC),它们的临床应用在很大程度上仍然受到获得性耐药介导的新EGFR突变和副作用的阻碍。蛋白水解靶向嵌合体(PROTAC)技术具有通过新的作用机制克服突变EGFR的获得性抗性的潜力。在这项研究中,我们通过先导化合物13的结构修饰开发了候选降解剂IV-3,其对HCC-827细胞表现出有限的抗增殖活性.与化合物13相比,IV-3对HCC-827细胞具有显著的抗增殖活性,NCI-H1975细胞,和NCI-H1975-TM细胞(IC50=0.009μM,0.49μM和3.24μM,分别),以及显着诱导这些细胞系中EGFR蛋白的降解(DC50=17.93nM,0.25μM和0.63μM,分别)。进一步的研究证实,通过突变EGFR蛋白的降解,IV-3在所有异种移植肿瘤模型中表现出优异的抗肿瘤活性。此外,IV-3对表达野生型EGFR的A431和A549细胞无抑制活性。从而消除了野生型EGFR抑制产生的潜在毒副作用。总的来说,我们的研究为EGFR-PROTACs作为EGFR获得性突变的潜在治疗策略提供了有希望的见解.
    Although three generations of Epidermal growth factor receptor (EGFR) - TK inhibitors have been approved for the treatment of Non-small-cell lung cancers (NSCLC), their clinical application is still largely hindered by acquired drug resistance mediated new EGFR mutations and side effects. The Proteolysis targeting chimera (PROTAC) technology has the potential to overcome acquired resistance from mutant EGFR through a novel mechanism of action. In this study, we developed the candidate degrader IV-3 by structural modifications of the lead compound 13, which exhibited limited antiproliferative activity against HCC-827 cells. Compared to compound 13, IV-3 exhibited remarkable anti-proliferative activity against HCC-827 cells, NCI-H1975 cells, and NCI-H1975-TM cells (IC50 = 0.009 μM, 0.49 μM and 3.24 μM, respectively), as well as significantly inducing degradation of EGFR protein in these cell lines (DC50 = 17.93 nM, 0.25 μM and 0.63 μM, respectively). Further investigations confirmed that IV-3 exhibited superior anti-tumor activity in all xenograft tumor models through the degradation of mutant EGFR protein. Moreover, IV-3 showed no inhibitory activity against A431 and A549 cells expressing wild-type EGFR, thereby eliminating potential toxic side effects emerging from wild-type EGFR inhibition. Overall, our study provides promising insights into EGFR-PROTACs as a potential therapeutic strategy against EGFR-acquired mutation.
    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
    癌症,以不受控制的细胞生长和转移为特征,造成近六分之一的死亡,对全球公共卫生构成严重威胁。化疗可以大大提高癌症患者的生活质量和生存率,但是抗癌化学疗法与一系列不良反应有关。此外,几乎所有目前可用的抗癌化学疗法都可以在癌症患者的一段时间内产生耐药性,并最终导致90%的患者癌症复发和死亡,迫切需要开发新的抗癌剂。融合嘧啶是DNA和RNA不可分割的部分,在许多生物过程中至关重要。融合嘧啶可以作用于各种生物癌症靶标,并具有解决耐药性的潜力。此外,超过20种融合嘧啶已经被批准用于不同癌症的临床治疗,并在当前的治疗武器库中占有重要地位,揭示了融合嘧啶是开发新型抗癌化学疗法的特权支架。这篇综述的目的是总结具有体内抗癌治疗潜力的融合嘧啶及其急性毒性的当前情况。代谢谱以及药代动力学特性,从2020年到现在发展起来的毒性和作用机制,以促进进一步合理利用更有效的候选物。
    Cancer, characterized by uncontrolled cell growth and metastasis, is responsible for nearly one in six deaths and represents a severe threat to public health worldwide. Chemotherapy can substantially improve the quality of life and survival of patients with cancer, but anticancer chemotherapeutics are associated with a range of adverse effects. Moreover, almost all currently available anticancer chemotherapeutics could develop drug resistance over a period of time of application in cancer patients and ultimately lead to cancer relapse and death in 90% of patients, creating an urgent need to develop new anticancer agents. Fused pyrimidines trait the inextricable part of DNA and RNA and are vital in numerous biological processes. Fused pyrimidines can act on various biological cancer targets and have the potential to address drug resistance. In addition, more than 20 fused pyrimidines have already been approved for clinical treatment of different cancers and occupy a prominent place in the current therapeutic arsenal, revealing that fused pyrimidines are privileged scaffolds for the development of novel anticancer chemotherapeutics. The purpose of this review is to summarize the current scenario of fused pyrimidines with in vivo anticancer therapeutic potential along with their acute toxicity, metabolic profiles as well as pharmacokinetic properties, toxicity and mechanisms of action developed from 2020 to the present to facilitate further rational exploitation of more effective candidates.
    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
    现代医学继续与抗生素抗性细菌病原体作斗争。在关键关注的病原体是多药耐药(MDR)铜绿假单胞菌,金黄色葡萄球菌,和肺炎克雷伯菌.这些病原体是免疫受损个体医院感染的主要原因,涉及肺等主要器官,皮肤,脾,脾肾,肝脏,和血液。因此,迫切需要新的方法。最近,我们开发了一种两亲性树枝状聚合物DDC18-8A,在体外具有高抗菌和抗生物膜功效。DDC18-8A由长的疏水烷基链和带有胺末端的小的亲水聚(酰胺基胺)树枝状体组成,通过将自身附着并插入细菌膜以触发细胞裂解来发挥其抗菌活性。这里,我们研究了DDC18-8A在人类传染病小鼠模型中的药代动力学和体内毒性以及抗菌功效。值得注意的是,DDC18-8A显着降低了铜绿假单胞菌急性肺炎和菌血症小鼠模型中的细菌负荷,耐甲氧西林金黄色葡萄球菌(MRSA),以及耐碳青霉烯类肺炎克雷伯菌和中性粒细胞减少性软组织感染铜绿假单胞菌和MRSA。最重要的是,DDC18-8A通过在低10倍的治疗浓度下实现相似的细菌清除而优于针对所有三种病原体的病原体特异性抗生素。此外,它在体内表现出优越的稳定性和生物分布,具有出色的安全性,但在主要器官的组织病理学分析中没有任何可观察到的异常,血清生物化学,和血液学。总的来说,我们提供了强有力的证据,证明DDC18-8A在解决与MDR病原体医院感染相关的挑战方面是目前处方抗生素的有希望的替代品.
    Modern medicine continues to struggle against antibiotic-resistant bacterial pathogens. Among the pathogens of critical concerns are the multidrug-resistant (MDR) Pseudomonas aeruginosa, Staphylococcus aureus, and Klebsiella pneumoniae. These pathogens are major causes of nosocomial infections among immunocompromised individuals, involving major organs such as lung, skin, spleen, kidney, liver, and bloodstream. Therefore, novel approaches are direly needed. Recently, we developed an amphiphilic dendrimer DDC18-8A exhibiting high antibacterial and antibiofilm efficacy in vitro. DDC18-8A is composed of a long hydrophobic alkyl chain and a small hydrophilic poly(amidoamine) dendron bearing amine terminals, exerting its antibacterial activity by attaching and inserting itself into bacterial membranes to trigger cell lysis. Here, we examined the pharmacokinetics and in vivo toxicity as well as the antibacterial efficacy of DDC18-8A in mouse models of human infectious diseases. Remarkably, DDC18-8A significantly reduced the bacterial burden in mouse models of acute pneumonia and bacteremia by P. aeruginosa, methicillin-resistant S. aureus (MRSA), and carbapenem-resistant K. pneumoniae and neutropenic soft tissue infection by P. aeruginosa and MRSA. Most importantly, DDC18-8A outperformed pathogen-specific antibiotics against all three pathogens by achieving a similar bacterial clearance at 10-fold lower therapeutic concentrations. In addition, it showed superior stability and biodistribution in vivo, with excellent safety profiles yet without any observable abnormalities in histopathological analysis of major organs, blood serum biochemistry, and hematology. Collectively, we provide strong evidence that DDC18-8A is a promising alternative to the currently prescribed antibiotics in addressing challenges associated with nosocomial infections by MDR pathogens.
    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
    多形性胶质母细胞瘤代表了实质性的临床挑战。瞬时受体电位通道(TRPC)拮抗剂可能为这种侵袭性癌症提供新的治疗选择。在这项研究中,使用支架跳跃策略设计并制备了一系列N-烷基-N-苯甲酰基和N-烷基-N-苄基噻唑,并将其作为TRPC6拮抗剂进行了评估.这导致了15g的发现,一种有效的TRPC拮抗剂,对TRPC3,TRPC4,TRPC5,TPRC6和TRPC7表现出合适的抑制性微摩尔活性,并在体外对U87细胞系表现出值得注意的抗胶质母细胞瘤功效。此外,15g具有可接受的药代动力学特征,并且在异种移植模型中表现出比一线治疗剂替莫唑胺(50mg/kg/d)更好的体内效力(25mg/kg/d)。一起来看,TRPC拮抗剂15g代表了开发新的抗胶质母细胞瘤药物的有前途的先导化合物。
    Glioblastoma multiforme represents a substantial clinical challenge. Transient receptor potential channel (TRPC) antagonists might provide new therapeutic options for this aggressive cancer. In this study, a series of N-alkyl-N-benzoyl and N-alkyl-N-benzyl thiazoles were designed and prepared using a scaffold-hopping strategy and evaluated as TRPC6 antagonists. This resulted in the discovery of 15g, a potent TRPC antagonist that exhibited suitable inhibitory micromolar activities against TRPC3, TRPC4, TRPC5, TPRC6, and TRPC7 and displayed noteworthy anti-glioblastoma efficacy in vitro against U87 cell lines. In addition, 15g featured an acceptable pharmacokinetic profile and exhibited better in vivo potency (25 mg/kg/d) than the frontline therapeutic agent temozolomide (50 mg/kg/d) in xenograft models. Taken together, the TRPC antagonist 15g represents a promising lead compound for developing new anti-glioblastoma agents.
    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
    化学疗法在不同类型癌症的药物治疗中占有举足轻重的作用,但是癌症的患病率和死亡率仍然很高。现有化疗药物的耐药性和低特异性是目前有效化疗的主要障碍,迫切需要开发新的抗癌剂。吡唑是一种具有两个相邻氮原子的高度通用的五元杂环,具有显着的治疗效果和强大的药理效力。吡唑衍生物,特别是吡唑杂化物已通过多种机制证明了有效的体外和体内抗癌功效。包括凋亡诱导,自噬调节,和细胞周期中断。此外,几种吡唑杂化物,如crizotanib(吡唑-吡啶杂化物),erdafitinib(吡唑-喹喔啉杂种)和ruxolitinib(吡唑-吡咯并[2,3-d]嘧啶杂种)已被批准用于癌症治疗,揭示吡唑杂种是开发新型抗癌剂的有用支架。这篇综述的目的是总结吡唑杂种的当前情况,具有潜在的体内抗癌功效以及作用机制。毒性,和药代动力学,涵盖近5年(2018年至今)发表的论文,促进进一步合理利用更有效的候选人。
    Chemotherapeutics occupy a pivotal role in the medication of different types of cancers, but the prevalence and mortality rates of cancer remain high. The drug resistance and low specificity of current available chemotherapeutics are the main barriers for the effective cancer chemotherapy, evoking an immediate need for the development of novel anticancer agents. Pyrazole is a highly versatile five-membered heterocycle with two adjacent nitrogen atoms and possesses remarkable therapeutic effects and robust pharmacological potency. The pyrazole derivatives especially pyrazole hybrids have demonstrated potent in vitro and in vivo efficacies against cancers through multiple mechanisms, inclusive of apoptosis induction, autophagy regulation, and cell cycle disruption. Moreover, several pyrazole hybrids such as crizotanib (pyrazole-pyridine hybrid), erdafitinib (pyrazole-quinoxaline hybrid) and ruxolitinib (pyrazole-pyrrolo [2,3-d]pyrimidine hybrid) have already been approved for the cancer therapy, revealing that pyrazole hybrids are useful scaffolds to develop novel anticancer agents. The purpose of this review is to summarize the current scenario of pyrazole hybrids with potential in vivo anticancer efficacy along with mechanisms of action, toxicity, and pharmacokinetics, covering papers published in recent 5 years (2018-present), to facilitate further rational exploitation of more effective candidates.
    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
    抗生素耐药性是一个不断升级的全球健康问题,未来每年可能导致数千万人死于耐药细菌感染。尤其是畜牧业。肽抗菌纳米材料由于其物理穿透致病性生物膜的独特机制,为抗生素提供了具有竞争力的替代品。本研究开发了具有高生物选择性的两亲性共组装肽纳米纤维(PCBP-NCAPNFs),以克服肽PCBP的高细胞毒性和肽NCAP的低抗菌活性。PCBP-NCAPNFs具有广谱抗菌活性和优异的生物相容性,体内和体外毒性可忽略不计。此外,使用仔猪全身感染模型,PCBP-NCAPNFs具有直接的抗菌功效和潜在的免疫调节能力。其独特的膜渗透机制和与病原菌表面阴离子成分结合的能力使其不易产生耐药性。总之,这些发现对于超分子肽纳米药物的临床应用和畜牧业的发展具有重要意义。
    Antibiotic resistance is an escalating global health concern that could result in tens of millions of deaths annually from drug-resistant bacterial infections in the future, especially in animal husbandry. Peptide antibacterial nanomaterials offer a competitive alternative to antibiotics because of their distinct mechanism of physically penetrating pathogenic biological membranes. This study developed amphiphilic co-assembled peptide nanofibers with high biological selectivity (PCBP-NCAP NFs) to overcome the high cytotoxicity of peptide PCBP and the low antibacterial activity of peptide NCAP. PCBP-NCAP NFs exhibit broad-spectrum antibacterial activity and excellent biocompatibility, with negligible in vivo and in vitro toxicity. Additionally, PCBP-NCAP NFs possess direct antibacterial efficacy and potential immunomodulatory capabilities using a piglet systemic infection model. Its unique mechanism of membrane penetration and the ability to bind to anionic components on the surface of pathogenic bacteria make them less susceptible to drug resistance. In conclusion, these findings have significant implications for the advancement of supramolecular peptide nanomedicines for clinical application and animal husbandry.
    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
    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
    1,2,3-Triazole moiety which is usually constructed by highly versatile, efficacious and selective copper-catalyzed azide-alkyne cycloaddition not only can act as a linker to connect different pharmacophores, but also is a useful pharmacophore with diverse biological properties. 1,2,3-Triazoles are readily interact with diverse enzymes and receptors in cancer cells through non-covalent interactions and can inhibit cancer cell proliferation, arrest cell cycle and induce apoptosis. In particular, 1,2,3-triazole-containing hybrids have the potential to exert dual or multiple anticancer mechanisms of action, representing useful scaffolds in expediting development of novel anticancer agents. The current review summarizes the in vivo anticancer efficacy and mechanisms of action of 1,2,3-triazole-containing hybrids reported in the last decade to continuously open up a map for the remarkable exploration of more effective candidates.
    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
    ALK是治疗非小细胞肺癌的一个有吸引力的治疗靶点。作为药物化学中的新兴元素,硼在发现抗肿瘤药物和抗菌药物方面取得了巨大的成功。通过BCC(含硼化合物)化合物库的构建和广泛的激酶筛选,我们发现ALK抑制剂击中化合物10a。通过CADD和等位排列进行结构优化表明,先导化合物10k具有改善的活性(ALKL1196MIC50=8.4nM,NCI-H2228细胞IC50=520nM)和更好的体外代谢稳定性(人肝微粒体,T1/2=238分钟)。化合物10k在TGI为52%的裸鼠NCI-H2228肺癌异种移植模型中显示出良好的体内功效。分子模拟分析结果表明,氧杂苯环上的羟基与Asn1254或Asp1270形成关键的氢键,该结合位点为药物设计提供了新思路。这是首次公开报道的含硼ALK抑制剂的先导化合物。
    ALK is an attractive therapeutic target for the treatment of non-small cell lung cancer. As an emerging element in medicinal chemistry, boron has achieved great success in the discovery of antitumor drugs and antibacterial agents. Through construction of a BCC (boron-containing compound) compound library and broad kinase screening, we found the ALK inhibitor hit compound 10a. Structural optimization by CADD and isosterism revealed that lead compound 10k has improved activity (ALKL1196M IC50 = 8.4 nM, NCI-H2228 cells IC50 = 520 nM) and better in vitro metabolic stability (human liver microsomes, T1/2 = 238 min). Compound 10k showed good in vivo efficacy in a nude mouse NCI-H2228 lung cancer xenograft model with a TGI of 52 %. Molecular simulation analysis results show that the hydroxyl group on the oxaborole forms a key hydrogen bond with Asn1254 or Asp1270, and this binding site provides a new idea for drug design. This is the first publicly reported lead compound for a boron-containing ALK inhibitor.
    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
    STAT3是治疗胃癌的一个有希望的治疗靶点,这是世界上最常见的实体瘤之一。在以前的作品中,我们发现了一系列带有咪唑并[1,2-a]吡啶支架的新型STAT3抑制剂。为了提高这些化合物的代谢稳定性,在本文中,我们进行了系统的结构优化,导致生物活性抑制剂42,其表现出对抑制生长的显着影响,人胃癌细胞系(AGS和MGC-803)的迁移和侵袭。同时,它能够在低微摩尔浓度下阻断STAT3的磷酸化和二聚化。此外,化合物42在MGC-803来源的异种移植小鼠模型中明显抑制肿瘤生长,这表明它作为一种有前途的晚期胃癌抗癌药物值得进一步探索。
    STAT3 is a promising therapeutic target for the treatment of gastric cancer, which is one of the most common solid tumors worldwide. In the previous works, we discovered a series of novel STAT3 inhibitors bearing an imidazo[1,2-a] pyridine scaffold. In order to improve the metabolic stability of these compounds, herein we performed a systematic structural optimization leading to a bioactive inhibitor 42, which demonstrated significant effects on inhibiting the growth, migration and invasion of human gastric cancer cells lines (AGS and MGC-803). Meanwhile, it was able to block the phosphorylation and dimerization of STAT3 at low micromolar concentration. Furthermore, compound 42 obviously suppressed tumor growth in MGC-803 derived xenograft mouse model, suggesting that it deserves further exploration as a promising anti-cancer agent for advanced gastric cancer.
    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