boceprevir

boceprevir
  • 文章类型: Journal Article
    虽然研究已经确定了SARS-CoV-2的主要蛋白酶(Mpro)的几种抑制剂,但这些化合物的很大一部分在还原剂存在下表现出降低的活性。引起人们对其体内有效性的担忧。此外,使用病毒颗粒的细胞试验的常规生物安全3级(BSL-3)对基于细胞的试验中Mpro抑制剂功效的广泛评估造成了限制.这里,我们建立了一个与BSL-1相容的细胞试验来评估Mpro抑制剂的体内潜力.该测定利用表达含有N-末端谷胱甘肽S-转移酶(GST)和C-末端血凝素(HA)标签的标记的Mpro构建体的哺乳动物细胞并监测Mpro自身消化。使用此方法,GC376和boceprevir有效抑制Mpro自身消化,表明它们在体内的潜在活性。相反,卡莫富和依布硒在该测定中没有表现出明显的抑制作用。我们使用这种方法进一步研究了硒酮对Mpro的抑制潜力。结合能的计算分析表明,非共价相互作用在促进C145残基的共价修饰中起关键作用。导致Mpro抑制。我们的方法很简单,成本效益高,并容易适用于标准实验室,使具有不同传染病专业知识水平的研究人员可以使用它。
    While research has identified several inhibitors of the main protease (Mpro) of SARS-CoV-2, a significant portion of these compounds exhibit reduced activity in the presence of reducing agents, raising concerns about their effectiveness in vivo. Furthermore, the conventional biosafety level 3 (BSL-3) for cellular assays using viral particles poses a limitation for the widespread evaluation of Mpro inhibitor efficacy in a cell-based assay. Here, we established a BSL-1 compatible cellular assay to evaluate the in vivo potential of Mpro inhibitors. This assay utilizes mammalian cells expressing a tagged Mpro construct containing N-terminal glutathione S-transferase (GST) and C-terminal hemagglutinin (HA) tags and monitors Mpro autodigestion. Using this method, GC376 and boceprevir effectively inhibited Mpro autodigestion, suggesting their potential in vivo activity. Conversely, carmofur and ebselen did not exhibit significant inhibitory effects in this assay. We further investigated the inhibitory potential of selenoneine on Mpro using this approach. Computational analyses of binding energies suggest that noncovalent interactions play a critical role in facilitating the covalent modification of the C145 residue, leading to Mpro inhibition. Our method is straightforward, cost-effective, and readily applicable in standard laboratories, making it accessible to researchers with varying levels of expertise in infectious diseases.
    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
    Boceprevir药物是一种酮酰胺丝氨酸蛋白酶抑制剂,具有线性拟肽结构,对2019-nCoV主要蛋白酶表现出抑制活性。本文报道了boceprevir的电子性质及其分子对接以及与蛋白质受体的分子动力学模拟分析。为此,通过DFT在6-311G的B3LYP和ωB97XD水平上获得了boceprevir的平衡结构(d,p)在气体和水介质中设置的基础。HOMO-LUMO和吸收光谱分析已用于评估beprevir的毒性和光敏性,分别。已经进行了分子对接模拟来测试boceprevir与2019-nCoVMPRO的结合亲和力;这在配体和靶蛋白的残基位置之间提供了多种理想的结合位置。最佳结合位置已被考虑用于分子动力学模拟。研究结果已得到解决,以阐明boceprevir药物对2019-nCoVMPRO的疗效。
    Boceprevir drug is a ketoamide serine protease inhibitor with a linear peptidomimetic structure that exhibits inhibition activity against 2019-nCoV main protease. This paper reports electronic properties of boceprevir and its molecular docking as well as molecular dynamics simulation analysis with protein receptor. For this, the equilibrium structure of boceprevir has been obtained by DFT at B3LYP and ωB97XD levels with 6-311+G(d,p) basis set in gas and water mediums. HOMO-LUMO and absorption spectrum analysis have been used to evaluate the boceprevir\'s toxicity and photosensitivity, respectively. Molecular docking simulation has been performed to test the binding affinity of boceprevir with 2019-nCoV MPRO; which rendered a variety of desirable binding locations between the ligand and target protein\'s residue positions. The optimum binding location has been considered for molecular dynamics simulation. The findings have been addressed to clarify the boceprevir drug efficacy against the 2019-nCoV MPRO.
    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: Review
    Nirmatrelvir,针对SARS-CoV-2主要蛋白酶(Mpro),是预防和治疗严重COVID-19的一线药物,其他Mpro抑制剂正在开发中。然而,耐药性发展的风险威胁到此类直接抗病毒药物的未来疗效.为了获得有关Mpro抑制剂耐药性的病毒相关知识,我们选择使用尼马特雷韦相关蛋白酶抑制剂boceprevir治疗的耐药SARS-CoV-2.在VeroE6细胞中进行的五次逃逸实验中选择的SARS-CoV-2显示出对尼马特雷韦的交叉抗性,与原始SARS-CoV-2相比,最大有效浓度的一半增加了7.3倍。序列分析显示逃逸病毒具有Mpro取代L50F和A173V。对于反向遗传研究,将这些替换引入细胞培养感染的SARS-CoV-2克隆中。细胞培养衍生的工程化SARS-CoV-2突变体的感染性滴定和遗传稳定性分析表明,L50F挽救了A173V赋予的适应性成本。在浓度反应实验中,A173V是对boceprevir和nirmatrelvir耐药的主要驱动因素。Mpro的结构分析表明,A173V可以通过使boceprevir和nirmatrelvir的结合不太有利而引起耐药性。这项研究有助于全面概述一线COVID-19治疗尼马特雷韦的耐药性,因此可以为人群监测提供信息,并有助于大流行的防范。
    Nirmatrelvir, which targets the SARS-CoV-2 main protease (Mpro), is the first-in-line drug for prevention and treatment of severe COVID-19, and additional Mpro inhibitors are in development. However, the risk of resistance development threatens the future efficacy of such direct-acting antivirals. To gain knowledge on viral correlates of resistance to Mpro inhibitors, we selected resistant SARS-CoV-2 under treatment with the nirmatrelvir-related protease inhibitor boceprevir. SARS-CoV-2 selected during five escape experiments in VeroE6 cells showed cross-resistance to nirmatrelvir with up to 7.3-fold increased half-maximal effective concentration compared to original SARS-CoV-2, determined in concentration-response experiments. Sequence analysis revealed that escape viruses harbored Mpro substitutions L50F and A173V. For reverse genetic studies, these substitutions were introduced into a cell-culture-infectious SARS-CoV-2 clone. Infectivity titration and analysis of genetic stability of cell-culture-derived engineered SARS-CoV-2 mutants showed that L50F rescued the fitness cost conferred by A173V. In the concentration-response experiments, A173V was the main driver of resistance to boceprevir and nirmatrelvir. Structural analysis of Mpro suggested that A173V can cause resistance by making boceprevir and nirmatrelvir binding less favorable. This study contributes to a comprehensive overview of the resistance profile of the first-in-line COVID-19 treatment nirmatrelvir and can thus inform population monitoring and contribute to pandemic preparedness.
    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
    背景:最近国际,随机化,安慰剂对照临床试验(SPRINT-2;RESPOND-2)表明,聚乙二醇干扰素(PEG),利巴韦林(RBV)和boceprevir(BOC)在治疗基因1型丙型肝炎病毒(HCV)慢性感染患者方面比标准的PEG和RBV双重治疗更有效.这项研究的目的是评估三联疗法在匈牙利初治和有治疗经验的患者中的成本效益。方法:从匈牙利付款人的角度开发了马尔可夫模型来评估三联疗法的长期临床益处和成本效益。模型状态为纤维化(F0-F4,使用METAVIR纤维化评分定义),失代偿期肝硬化(DC),肝细胞癌(HCC),肝移植,和肝脏相关死亡(LD)。根据SPRINT-2和RESPOND-2研究估计疗效。模型中使用的疾病进展率和健康状况效用是从已发表的研究中获得的。肝移植的可能性和成本的估计是基于对匈牙利疾病基金数据库的分析。所有成本和收益都以每年5%的价格打折。结果:与双重疗法相比,三联疗法预计将增加预期寿命0.98和2.42寿命年,并增加质量调整寿命年(QALY)0.59和1.13治疗初治和治疗经验丰富的患者,分别。相应的增量成本效益比分别为每QALYHUF7,747,962(26,717欧元)和HUF5,888,240(20,304欧元)。严重肝病事件的终生发生率(DC,HCC,LT,与PEG-RBV治疗相比,在接受三联疗法治疗的未治疗和有治疗经验的患者中,LD)预计将降低45%和61%。结论:在匈牙利,将boceprevir添加到治疗1型慢性HCV感染患者的标准疗法中,使用常用的支付意愿阈值为846万HUF(人均国内生产总值的3倍),预计具有成本效益。
    Background: Recent international, randomized, placebo-controlled clinical trials (SPRINT-2; RESPOND-2) demonstrated that the triple combination of peginterferon (PEG), ribavirin (RBV) and boceprevir (BOC) was more efficacious than the standard dual therapy of PEG and RBV in treatment of patients chronically infected with genotype 1 hepatitis C virus (HCV) infection. The objective of this study was to evaluate the cost-effectiveness of triple therapy in both treatment-naive and treatment-experienced patients in Hungary. Methods: A Markov model was developed to evaluate the long-term clinical benefits and the costeffectiveness of the triple therapy from the Hungarian payer perspective. Model states were fibrosis (F0-F4, defined using METAVIR fibrosis scores), decompensated cirrhosis (DC), hepatocellular carcinoma (HCC), liver transplantation (LT), and liver-related deaths (LD). Efficacy was estimated from SPRINT-2 and RESPOND-2 studies. Disease progression rates and health state utilities used in the model were obtained from published studies. Estimates of probability of liver transplantation and cost were based on an analysis of the Hungarian Sick Fund database. All cost and benefits were discounted at 5% per year. Results: Compared to dual therapy, triple therapy was projected to increase the life expectancy by 0.98 and 2.42 life years and increase the quality-adjusted life years (QALY) by 0.59 and 1.13 in treatment-naive and treatment-experienced patients, respectively. The corresponding incremental cost-effectiveness ratios were HUF7,747,962 (€26,717) and HUF5,888,240 (€20,304) per QALY. The lifetime incidence of severe liver disease events (DC, HCC, LT, LD) were projected to decrease by 45% and 61% in treatment-naïve and treatment-experienced patients treated with triple therapy groups in comparison with PEG-RBV treatment. Conclusion: The addition of boceprevir to standard therapy for the treatment of patients with genotype 1 chronic HCV infection in Hungary is projected to be cost-effective using a commonly used willingness to pay threshold of HUF 8.46 million (3 times gross domestic product per capita).
    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
    近年来,由于SARS-CoV-2病毒引起的COVID-19流行病学疾病,世界上200多个国家面临着健康危机。它对世界经济和全球卫生部门产生了巨大影响。研究人员正在研究可以抑制SARS-CoV-2的药物的设计和发现。SARS-CoV-2的主要蛋白酶是研究针对冠状病毒疾病的抗病毒药物的有吸引力的目标。根据对接结果,Boceprevir的结合能,马赛替尼和鲁普莱维与CMP分别为-10.80、-9.39和-9.51kcal/mol。此外,对于所有被调查的系统,范德华和静电相互作用非常有利于药物与SARS-CoV-2冠状病毒主要蛋白酶的结合,表明复杂稳定性的确认。
    In recent years, more than 200 countries of the world have faced a health crisis due to the epidemiological disease of COVID-19 caused by the SARS-CoV-2 virus. It had a huge impact on the world economy and the global health sector. Researchers are studying the design and discovery of drugs that can inhibit SARS-CoV-2. The main protease of SARS-CoV-2 is an attractive target for the study of antiviral drugs against coronavirus diseases. According to the docking results, binding energy for boceprevir, masitinib and rupintrivir with CMP are -10.80, -9.39, and -9.51 kcal/mol respectively. Also, for all investigated systems, van der Waals and electrostatic interactions are quite favorable for binding the drugs to SARS-CoV-2 coronavirus main protease, indicating confirmation of the complex stability.
    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
    背景:无法获得针对SARS-CoV-2病毒感染的靶特异性抗病毒药物,激发了将6,6-二甲基-3-氮杂双环[3.1.0]己烷-2-甲酰胺的衍生物设计为针对相关病毒的潜在抗病毒抑制剂的动机。分子对接和分子动力学结果表明,所报道的衍生物具有作为抗SARS-CoV-2抗病毒药物的潜力。所报道的命中化合物可考虑用于体外和体内分析。
    方法:使用基于片段的药物设计来模拟衍生物。此外,使用B3LYP/6-311G**基集进行DFT模拟。对接模拟是通过在AutoDock4.2下使用经验自由能力场与拉马克遗传算法的组合进行的。通过应用AMBER14力场和SPCE水模型,分子动力学模拟和MM-PBSA计算为100ns。
    BACKGROUND: The unavailability of target-specific antiviral drugs for SARS-CoV-2 viral infection kindled the motivation to virtually design derivatives of 6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide as potential antiviral inhibitors against the concerned virus. The molecular docking and molecular dynamic results revealed that the reported derivatives have a potential to act as antiviral drug against SARS-CoV-2. The reported hit compounds can be considered for in vitro and in vivo analyses.
    METHODS: Fragment-based drug designing was used to model the derivatives. Furthermore, DFT simulations were carried out using B3LYP/6-311G** basis set. Docking simulations were performed by using a combination of empirical free energy force field with a Lamarckian genetic algorithm under AutoDock 4.2. By the application of AMBER14 force field and SPCE water model, molecular dynamic simulations and MM-PBSA were calculated for 100 ns.
    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
    β-冠状病毒SARS-CoV-2的主要蛋白酶(Mpro)是开发COVID-19治疗方法的有吸引力的靶标。基于结构的设计是发现Mpro的新抑制剂的成功方法。从与丙型肝炎病毒NS3/4A蛋白酶抑制剂boceprevir和telaprevir复合物中的Mpro的晶体结构开始,我们通过用γ-内酰胺替代其P1环丁基部分作为谷氨酰胺替代品,优化了α-酮酰胺boceprevir对Mpro的效力.生成的化合物,与重组Mpro相比,MG-78表现出13nM的IC50,其P1'N-甲基衍生物MG-131的效力相似。晶体结构证实了我们设计概念的有效性。除了SARS-CoV-2Mpro抑制,我们还探讨了MG-78对α冠状病毒HCoVNL63的Mpro和肠道病毒3C蛋白酶的活性。活动良好(0.33µM,HCoV-NL63Mpro),中等(1.45µM,柯萨奇病毒3Cpro),和相对较差(6.7µM,肠道病毒A713Cpro),分别。通过X射线晶体学揭示了活性差异的结构基础。我们得出的结论是,改良的boceprevir支架适用于获得冠状病毒Mpros的高效抑制剂,但需要进一步优化才能有效地靶向肠道病毒3Cpros。
    The main protease (Mpro) of the betacoronavirus SARS-CoV-2 is an attractive target for the development of treatments for COVID-19. Structure-based design is a successful approach to discovering new inhibitors of the Mpro. Starting from crystal structures of the Mpro in complexes with the Hepatitis C virus NS3/4A protease inhibitors boceprevir and telaprevir, we optimized the potency of the alpha-ketoamide boceprevir against the Mpro by replacing its P1 cyclobutyl moiety by a γ-lactam as a glutamine surrogate. The resulting compound, MG-78, exhibited an IC50 of 13 nM versus the recombinant Mpro, and similar potency was observed for its P1\' N-methyl derivative MG-131. Crystal structures confirmed the validity of our design concept. In addition to SARS-CoV-2 Mpro inhibition, we also explored the activity of MG-78 against the Mpro of the alphacoronavirus HCoV NL63 and against enterovirus 3C proteases. The activities were good (0.33 µM, HCoV-NL63 Mpro), moderate (1.45 µM, Coxsackievirus 3Cpro), and relatively poor (6.7 µM, enterovirus A71 3Cpro), respectively. The structural basis for the differences in activities was revealed by X-ray crystallo-graphy. We conclude that the modified boceprevir scaffold is suitable for obtaining high-potency inhibitors of the coronavirus Mpros but further optimization would be needed to target enterovirus 3Cpros efficiently.
    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
    新型冠状病毒病(COVID-19)已在全球蔓延,影响数百万人。世界卫生组织(WHO)已宣布这种传染病为大流行。目前,几项临床试验正在进行中,以确定治疗这种感染的可能药物。SARS-CoV-2Mpro是阻断病毒复制的最关键的药物靶标之一。这项研究的目的是鉴定可能与SARS-CoV-2主要蛋白酶的活性位点结合并阻止病毒复制的潜在天然蒽醌。使用SwissDOCK服务器进行了13种蒽醌和一种对照药物(Boceprevir)与SARS-CoV-2Mpro的盲分子对接研究,和对Mpro表现出最高结合亲和力的alterporriol-Q进行了分子动力学模拟研究。这项研究表明,几种抗病毒蒽醌可以证明是有效的抑制SARS-CoV-2MPro的COVID-19,因为它们结合在具有催化二元的活性位点附近,HIS41和CYS145通过非共价力。与FDA批准的药物相比,蒽醌的抑制潜力较小,boceprevir.在研究的蒽醌中,发现alterporriol-Q是SARS-CoV-2Mpro的最有效抑制剂。Further,对Mpro-alterporriol-Q系统的MD模拟研究表明,alterporriol-Q不会在很大程度上改变Mpro的结构。考虑到COVID-19的影响,鉴定出可以抑制病毒感染的替代化合物,如alterporriol-Q,将有助于加速药物发现的过程。
    UNASSIGNED:在线版本包含补充材料,可在10.1007/s11756-021-01004-4获得。
    The novel coronavirus disease (COVID-19) has spread throughout the globe, affecting millions of people. The World Health Organization (WHO) has declared this infectious disease a pandemic. At present, several clinical trials are going on to identify possible drugs for treating this infection. SARS-CoV-2 Mpro is one of the most critical drug targets for the blockage of viral replication. The aim of this study was to identify potential natural anthraquinones that could bind to the active site of SARS-CoV-2 main protease and stop the viral replication. Blind molecular docking studies of 13 anthraquinones and one control drug (Boceprevir) with SARS-CoV-2 Mpro were carried out using the SwissDOCK server, and alterporriol-Q that showed the highest binding affinity towards Mpro were subjected to molecular dynamics simulation studies. This study indicated that several antiviral anthraquinones could prove to be effective inhibitors for SARS-CoV-2 Mpro of COVID-19 as they bind near the active site having the catalytic dyad, HIS41 and CYS145 through non-covalent forces. The anthraquinones showed less inhibitory potential as compared to the FDA-approved drug, boceprevir. Among the anthraquinones studied, alterporriol-Q was found to be the most potent inhibitor of SARS-CoV-2 Mpro. Further, MD simulation studies for Mpro- alterporriol-Q system suggested that alterporriol-Q does not alter the structure of Mpro to a significant extent. Considering the impact of COVID-19, identification of alternate compounds like alterporriol-Q that could inhibit the viral infection will help in accelerating the process of drug discovery.
    UNASSIGNED: The online version contains supplementary material available at 10.1007/s11756-021-01004-4.
    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
    Because of the scale of the novel coronavirus (COVID-19) pandemic and the swift transmission of this highly contagious respiratory virus, repurposing existing drugs has become an urgent treatment approach. The objective of our study is to unravel the binding mechanism of the Food and Drug Administration (FDA)-approved dexamethasone (Dex) and boceprevir (Boc) drugs with selected COVID-19 protein targets SARS-CoV-2 spike protein C-terminal domain (spike-CTD), main protease (Mpro), and interleukin-6 (IL-6). Another objective is to analyze the effects of binding Dex and Boc drugs on the interactions of viral spike protein to human angiotensin-converting enzyme 2 (hACE2). Molecular docking and one-microsecond-long molecular dynamics simulations of each of the six protein-drug complexes along with steered molecular dynamics (SMD) and umbrella sampling (US) methods have revealed the binding mode interactions and the physicochemical stability of the three targeted proteins with two drugs. Results have shown that both drugs bind strongly with the three protein targets through hydrogen bonding and hydrophobic interactions. A major finding from this study is how the binding of the drugs with viral spike protein affects its interactions at the binding interface with hACE2 protein. Simulations of drug-bound spike-CTD with hACE2 show that due to the presence of a drug at the binding interface of spike-CTD, hACE2 is being blocked from making putative interactions with viral protein at such interface. These important findings regarding the binding affinity and stability of the two FDA-approved drugs with the main targets of COVID-19 along with the effect of drugs on hACE2 interactions would contribute to COVID-19 drug discovery and development.
    UNASSIGNED: The online version contains supplementary material available at 10.1007/s11696-021-01843-0.
    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
    Remdesivir(GS-5734),一种最初开发用于治疗丙型肝炎和埃博拉病毒病的药物,是2019年首个批准的严重冠状病毒病治疗方案(COVID-19)。然而,除了Remdesivir,缺乏其他抗SARS-CoV-2感染的特异性抗病毒药物。2017年,研究人员在动物模型中记录了remdesivir的抗冠状病毒潜力。同时,在非洲两次埃博拉疫情期间进行的试验表明,这种药物是安全的。尽管针对SARS-CoV-2感染的疫苗已经以非常高的速度出现,抗病毒药物开发的努力取得了同等的结果,在大流行的现阶段,这本来可以发挥真正的拯救生命的作用,一直停滞不前。在这次审查中,我们将重点关注COVID-19的当前治疗方案,这些方案主要包括再利用的药物或赋予被动免疫的治疗方案(恢复期血浆或单克隆抗体)。此外,将审查正在开发的潜在特异性抗病毒疗法,如诱饵小蛋白CTC-445.2d,蛋白酶抑制剂,主要针对主要蛋白质Mpro,核苷类似物,如molnupiravir和化合物阻断复制转录复合蛋白,比如zotatifin和plitidepsin.这些抗病毒剂似乎是非常有前途的,但仍需要细致的临床试验测试,以确定其疗效和安全性。病毒变体的不断出现可能对科学界提出了真正的挑战。在这种情况下,纳米抗体的出现以及抗病毒药物组合的潜在给药可作为对抗COVID-19的医疗设备的有用工具.
    Remdesivir (GS-5734), a drug initially developed to treat hepatitis C and Ebola virus disease, was the first approved treatment for severe coronavirus disease 2019 (COVID-19). However, apart from remdesivir, there is a paucity of other specific anti-viral agents against SARS-CoV-2 infection. In 2017, researchers had documented the anti-coronavirus potential of remdesivir in animal models. At the same time, trials performed during two Ebola outbreaks in Africa showed that the drug was safe. Although vaccines against SARS-CoV-2 infection have emerged at an enormously high speed, equivalent results from efforts towards the development of anti-viral drugs, which could have played a truly life-saving role in the current stage of the pandemic, have been stagnating. In this review, we will focus on the current treatment options for COVID-19 which mainly consist of repurposed agents or treatments conferring passive immunity (convalescent plasma or monoclonal antibodies). Additionally, potential specific anti-viral therapies under development will be reviewed, such as the decoy miniprotein CTC-445.2d, protease inhibitors, mainly against the Main protein Mpro, nucleoside analogs, such as molnupiravir and compounds blocking the replication transcription complex proteins, such as zotatifin and plitidepsin. These anti-viral agents seem to be very promising but still require meticulous clinical trial testing in order to establish their efficacy and safety. The continuous emergence of viral variants may pose a real challenge to the scientific community towards that end. In this context, the advent of nanobodies together with the potential administration of a combination of anti-viral drugs could serve as useful tools in the armamentarium against COVID-19.
    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