Evaluation of potential anti-COVID-19 therapies

Clare L Box, Kevin S J Thompson
{"title":"Evaluation of potential anti-COVID-19 therapies","authors":"Clare L Box, Kevin S J Thompson","doi":"10.4155/fdd-2020-0029","DOIUrl":null,"url":null,"abstract":"COVID-19 – the infectious disease caused by the SARS-CoV-2 virus, a member of the ssRNA coronavirus family – has given rise to over 51.5 million confirmed infections and 1,275,979 deaths worldwide, as of 12 November 2020 [1]. It is known to be related to both the SARS-CoV and MERS-CoV viruses, responsible for severe acute respiratory syndrome and Middle East respiratory syndrome, respectively [2–4]. Human-to-human transmission of COVID-19 is widely recognized to be through a respiratory mechanism [3]. The severity of the disease ranges from asymptomatic to fatal [4,5]. The main reported symptoms include fever, nonproductive cough and loss of taste and smell, with severe cases reporting acute respiratory distress, viral pneumonia and requiring intubation and mechanical ventilation [1,4,6]. To date, there is no clinically approved vaccine available, nor any antiviral drug treatment for severe cases of this disease [1,4].\n Virus-encoded anti-infective targets\n A number of potential drug targets expressed by members of the coronavirus family have been identified. These include the ‘Spike’ glycoprotein, a trimer that binds to ACE2 on the host cell membrane [3,4]. This allows fusion of the viral and host cell membranes and viral entry into the cell [3]. Other potential targets include the cysteine proteases – 3CLpro (also known as Mpro) and PLpro – which are essential for production of new mature virions [7,8]. Another potential target is RdRP, which is needed for replication of the viral genome [5].\n Drug repurposing\n In order to rapidly progress new drug therapies into clinical use against COVID-19, drug repurposing has been widely investigated [9]. This has the advantage that potential therapeutics have already been approved for use in humans [9]. Virtual screening has been widely employed to aid in the repurposing of existing drug therapies for COVID-19 [2,10]. This approach is more rapid and economical than conventional lab-based testing. A number of potential drug treatments have been identified using this process. These include remdesivir, previously used for treatment of the Ebola and hepatitis C viruses and ribavirin, previously used for treatment of respiratory syncytial virus infection, hepatitis C and some hemorrhagic fevers. Remdesivir is reported to target RdRP, whereas ribavirin has been reported to target both 3CLpro and RdRP [2,6,11]. Recently, the US FDA (MD, USA) approved the use of remdesivir for COVID-19 patients aged over 12 years [12]. However, the WHO (Geneva, Switzerland) have released unpublished data from a clinical trial suggesting remdesivir does not reduce the mortality rate, shorten hospital stays or reduce the need for ventilation in patients with severe COVID-19 [1]. Other clinical trials of drug combinations are still ongoing.\n Development of new therapeutic antiviral agents\n The need to develop novel, targeted antivirals to treat SARS-CoV-2 infection is clear. However, the drug discovery and development process is likely too lengthy to address the current pandemic. A fundamental understanding of the structure–activity relationship of any potential new therapeutic for its SARS-CoV-2 target is essential. There is high amino acid sequence homology between coronaviruses. For instance, SARS-CoV-2 Spike glycoprotein and 3CLpro share 76 and 96% amino acid sequence homology, respectively, with those of SARS-CoV [2,10]. Where sequence homology is high and protein structure is similar, it is likely that inhibitors developed against SARS-CoV will be effective against SARS-CoV-2 [5,9]. If not, such compounds are likely at least to provide good leads for optimization and subsequent drug development [5,9]. Computational modeling and virtual screening may assist to quickly identify leads, backed up by lab-based functional testing and x-ray crystallography to generate novel structure–activity relationship for further lead optimization [2].\n When designing assays to test potential therapeutics, it is important to consider the functional state of the target protein. For instance, 3CLpro from SARS-CoV has been found only to be active as a dimer in solution [13]. It has also been demonstrated that additional affinity tags at either the C- or N-terminus are likely to reduce its enzymatic activity [13]. Due to the high sequence homology with 3CLpro from SARS-CoV-2 these criteria should be examined when establishing suitable screening assays [2,13]. A FRET-based fluorescent substrate assay and unlabeled 3CLpro has been used to determine the IC50 of potential drug-like ligands targeting SARS-CoV 3CLpro [8,13]. Similarly, isothermal titration calorimetry (ITC) has been used to determine the binding affinity of potential drugs [8].\n Host-encoded therapeutic targets\n ACE2, expressed on lung alveolar epithelial cells (as well as in the heart, kidney and testes) is the target for SARS-Cov-2 Spike glycoprotein binding [3]. This interaction allows SARS-CoV-2 membrane fusion with the host cell and entry of the viral genome [3]. A possible therapeutic approach has been trialed using a recombinant soluble form of human ACE2 to block Spike binding to ACE2 expressed on cell surfaces [14]. This prevents viral entry into the host cell, blocking viral replication and reducing viral burden [3,14]. Our unpublished observations using the label-free, surface plasmon resonance (SPR) technique to analyze the biomolecular interaction between ACE2 and Spike glycoprotein elegantly demonstrates this. Spike glycoprotein pre-incubated with soluble ACE2 markedly reduces Spike binding to ACE2 immobilized onto the SPR sensor surface [15].\n Anti-inflammatory drugs\n Treatments for COVID-19 are not limited to antivirals. They also include the use of drugs to reduce host immune responses. One documented issue is an increase in Ang II that, in addition to its cardiovascular role, acts as an inflammatory protein [16]. Ang II is degraded by ACE2 [16]. Endocytosis of the Spike-ACE2 complex, the mechanism by which SARS-CoV-2 enters cells, is thought to lead to a reduction in ACE2 availability which, in turn, contributes to an increase in Ang II. High levels of Ang II binding to AT1 lead to a signalling cascade which results in pro-inflammatory responses [16]. This in turn, may lead to acute respiratory distress [16]. It has been proposed that AT1 blockers could be used to compete with Ang II for AT1 binding sites [16]. The affinity for various AT1 blockers has been measured using radioligand binding assays [16] – another useful tool in the drug discovery process.\n Other approaches to reduce the inflammatory response include treatment with corticosteroids such as dexamethasone, which reduces death by up to a third in patients on ventilators [17]. Such treatments are useful for severely affected patients to reduce side effects. However, corticosteroids have no antiviral properties and are not recommended for treatment of early stage disease [17].\n Antibody-based therapies\n Infusion of plasma from recovered, post-COVID-19 infection donors, containing antibodies against SARS-CoV-2 antigens, has been used prophylactically to prevent infection in high-risk individuals [18,19]. However, plasma from a single postinfection donor is only sufficient for up to three recipients. Use of recombinant monoclonal antibodies targeting the viral Spike glycoprotein and manufactured in bulk quantities overcomes the limited supply of postinfection plasma [18,19]. Clinical trials of monoclonal antibodies in patients with severe disease are ongoing, but an effective treatment has not yet been approved [1,18]. All such protein infusion therapies (antibodies and soluble ACE2) are limited by the degradation of the protein by the host and require regular retreatment to maintain protection.\n Vaccination approaches\n As with most viral diseases, preventing establishment of the infection in the host and the subsequent spread of the virus by a global program of vaccination is regarded as the most successful approach. While approval of an effective vaccine is awaited, many countries are implementing social distancing and the use of face coverings to control the transmission of the virus [4]. The need for an effective vaccine is clear. Over 150 vaccine trials are reported to be underway [1,4]. Most vaccines target the SARS-CoV-2 Spike glycoprotein [4]. A variety of types are currently undergoing various stages of clinical trials including protein subunit vaccines, RNA-based vaccines and replicating viral vector vaccines [4].\n Pfizer (NY, USA) and BioNTech (Mainz, Germany) recently announced that their codon-optimized mRNA vaccine (BNT162b1) Phase III trial was 90% effective at protecting against COVID-19 infection [20]. However, the data have not been subjected to peer review. An effective vaccine would help to prevent the spread of the disease, allowing social distancing measures to be eased and a return toward social normality.\n Conclusion\n While drug repurposing has shown some limited success, it is clear that a targeted antiviral drug is required to treat patients with ongoing SARS-CoV-2 infection. A vaccine against SARS-CoV-2 is most likely to prevent infection from establishing within the host and hence the further spread of the virus, reducing mortality worldwide.","PeriodicalId":73122,"journal":{"name":"Future drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2020-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.4155/fdd-2020-0029","citationCount":"1","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Future drug discovery","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.4155/fdd-2020-0029","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 1

Abstract

COVID-19 – the infectious disease caused by the SARS-CoV-2 virus, a member of the ssRNA coronavirus family – has given rise to over 51.5 million confirmed infections and 1,275,979 deaths worldwide, as of 12 November 2020 [1]. It is known to be related to both the SARS-CoV and MERS-CoV viruses, responsible for severe acute respiratory syndrome and Middle East respiratory syndrome, respectively [2–4]. Human-to-human transmission of COVID-19 is widely recognized to be through a respiratory mechanism [3]. The severity of the disease ranges from asymptomatic to fatal [4,5]. The main reported symptoms include fever, nonproductive cough and loss of taste and smell, with severe cases reporting acute respiratory distress, viral pneumonia and requiring intubation and mechanical ventilation [1,4,6]. To date, there is no clinically approved vaccine available, nor any antiviral drug treatment for severe cases of this disease [1,4]. Virus-encoded anti-infective targets A number of potential drug targets expressed by members of the coronavirus family have been identified. These include the ‘Spike’ glycoprotein, a trimer that binds to ACE2 on the host cell membrane [3,4]. This allows fusion of the viral and host cell membranes and viral entry into the cell [3]. Other potential targets include the cysteine proteases – 3CLpro (also known as Mpro) and PLpro – which are essential for production of new mature virions [7,8]. Another potential target is RdRP, which is needed for replication of the viral genome [5]. Drug repurposing In order to rapidly progress new drug therapies into clinical use against COVID-19, drug repurposing has been widely investigated [9]. This has the advantage that potential therapeutics have already been approved for use in humans [9]. Virtual screening has been widely employed to aid in the repurposing of existing drug therapies for COVID-19 [2,10]. This approach is more rapid and economical than conventional lab-based testing. A number of potential drug treatments have been identified using this process. These include remdesivir, previously used for treatment of the Ebola and hepatitis C viruses and ribavirin, previously used for treatment of respiratory syncytial virus infection, hepatitis C and some hemorrhagic fevers. Remdesivir is reported to target RdRP, whereas ribavirin has been reported to target both 3CLpro and RdRP [2,6,11]. Recently, the US FDA (MD, USA) approved the use of remdesivir for COVID-19 patients aged over 12 years [12]. However, the WHO (Geneva, Switzerland) have released unpublished data from a clinical trial suggesting remdesivir does not reduce the mortality rate, shorten hospital stays or reduce the need for ventilation in patients with severe COVID-19 [1]. Other clinical trials of drug combinations are still ongoing. Development of new therapeutic antiviral agents The need to develop novel, targeted antivirals to treat SARS-CoV-2 infection is clear. However, the drug discovery and development process is likely too lengthy to address the current pandemic. A fundamental understanding of the structure–activity relationship of any potential new therapeutic for its SARS-CoV-2 target is essential. There is high amino acid sequence homology between coronaviruses. For instance, SARS-CoV-2 Spike glycoprotein and 3CLpro share 76 and 96% amino acid sequence homology, respectively, with those of SARS-CoV [2,10]. Where sequence homology is high and protein structure is similar, it is likely that inhibitors developed against SARS-CoV will be effective against SARS-CoV-2 [5,9]. If not, such compounds are likely at least to provide good leads for optimization and subsequent drug development [5,9]. Computational modeling and virtual screening may assist to quickly identify leads, backed up by lab-based functional testing and x-ray crystallography to generate novel structure–activity relationship for further lead optimization [2]. When designing assays to test potential therapeutics, it is important to consider the functional state of the target protein. For instance, 3CLpro from SARS-CoV has been found only to be active as a dimer in solution [13]. It has also been demonstrated that additional affinity tags at either the C- or N-terminus are likely to reduce its enzymatic activity [13]. Due to the high sequence homology with 3CLpro from SARS-CoV-2 these criteria should be examined when establishing suitable screening assays [2,13]. A FRET-based fluorescent substrate assay and unlabeled 3CLpro has been used to determine the IC50 of potential drug-like ligands targeting SARS-CoV 3CLpro [8,13]. Similarly, isothermal titration calorimetry (ITC) has been used to determine the binding affinity of potential drugs [8]. Host-encoded therapeutic targets ACE2, expressed on lung alveolar epithelial cells (as well as in the heart, kidney and testes) is the target for SARS-Cov-2 Spike glycoprotein binding [3]. This interaction allows SARS-CoV-2 membrane fusion with the host cell and entry of the viral genome [3]. A possible therapeutic approach has been trialed using a recombinant soluble form of human ACE2 to block Spike binding to ACE2 expressed on cell surfaces [14]. This prevents viral entry into the host cell, blocking viral replication and reducing viral burden [3,14]. Our unpublished observations using the label-free, surface plasmon resonance (SPR) technique to analyze the biomolecular interaction between ACE2 and Spike glycoprotein elegantly demonstrates this. Spike glycoprotein pre-incubated with soluble ACE2 markedly reduces Spike binding to ACE2 immobilized onto the SPR sensor surface [15]. Anti-inflammatory drugs Treatments for COVID-19 are not limited to antivirals. They also include the use of drugs to reduce host immune responses. One documented issue is an increase in Ang II that, in addition to its cardiovascular role, acts as an inflammatory protein [16]. Ang II is degraded by ACE2 [16]. Endocytosis of the Spike-ACE2 complex, the mechanism by which SARS-CoV-2 enters cells, is thought to lead to a reduction in ACE2 availability which, in turn, contributes to an increase in Ang II. High levels of Ang II binding to AT1 lead to a signalling cascade which results in pro-inflammatory responses [16]. This in turn, may lead to acute respiratory distress [16]. It has been proposed that AT1 blockers could be used to compete with Ang II for AT1 binding sites [16]. The affinity for various AT1 blockers has been measured using radioligand binding assays [16] – another useful tool in the drug discovery process. Other approaches to reduce the inflammatory response include treatment with corticosteroids such as dexamethasone, which reduces death by up to a third in patients on ventilators [17]. Such treatments are useful for severely affected patients to reduce side effects. However, corticosteroids have no antiviral properties and are not recommended for treatment of early stage disease [17]. Antibody-based therapies Infusion of plasma from recovered, post-COVID-19 infection donors, containing antibodies against SARS-CoV-2 antigens, has been used prophylactically to prevent infection in high-risk individuals [18,19]. However, plasma from a single postinfection donor is only sufficient for up to three recipients. Use of recombinant monoclonal antibodies targeting the viral Spike glycoprotein and manufactured in bulk quantities overcomes the limited supply of postinfection plasma [18,19]. Clinical trials of monoclonal antibodies in patients with severe disease are ongoing, but an effective treatment has not yet been approved [1,18]. All such protein infusion therapies (antibodies and soluble ACE2) are limited by the degradation of the protein by the host and require regular retreatment to maintain protection. Vaccination approaches As with most viral diseases, preventing establishment of the infection in the host and the subsequent spread of the virus by a global program of vaccination is regarded as the most successful approach. While approval of an effective vaccine is awaited, many countries are implementing social distancing and the use of face coverings to control the transmission of the virus [4]. The need for an effective vaccine is clear. Over 150 vaccine trials are reported to be underway [1,4]. Most vaccines target the SARS-CoV-2 Spike glycoprotein [4]. A variety of types are currently undergoing various stages of clinical trials including protein subunit vaccines, RNA-based vaccines and replicating viral vector vaccines [4]. Pfizer (NY, USA) and BioNTech (Mainz, Germany) recently announced that their codon-optimized mRNA vaccine (BNT162b1) Phase III trial was 90% effective at protecting against COVID-19 infection [20]. However, the data have not been subjected to peer review. An effective vaccine would help to prevent the spread of the disease, allowing social distancing measures to be eased and a return toward social normality. Conclusion While drug repurposing has shown some limited success, it is clear that a targeted antiviral drug is required to treat patients with ongoing SARS-CoV-2 infection. A vaccine against SARS-CoV-2 is most likely to prevent infection from establishing within the host and hence the further spread of the virus, reducing mortality worldwide.
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
评估潜在的抗covid -19疗法
COVID-19是由ssRNA冠状病毒家族成员SARS-CoV-2病毒引起的传染病,截至2020年11月12日,全球已造成5150多万例确诊感染和1,275,979例死亡。已知它与SARS-CoV和MERS-CoV病毒都有关系,这两种病毒分别导致严重急性呼吸综合征和中东呼吸综合征[2-4]。COVID-19的人际传播被广泛认为是通过呼吸机制进行的。该病的严重程度从无症状到致命不等[4,5]。报告的主要症状包括发热、无反应性咳嗽和味觉和嗅觉丧失,严重者报告急性呼吸窘迫、病毒性肺炎并需要插管和机械通气[1,4,6]。迄今为止,还没有临床批准的疫苗,也没有针对该病重症病例的抗病毒药物治疗[1,4]。由冠状病毒家族成员表达的许多潜在药物靶点已被确定。其中包括“Spike”糖蛋白,这是一种与宿主细胞膜上的ACE2结合的三聚体[3,4]。这使得病毒和宿主细胞膜融合,病毒进入细胞[3]。其他潜在的靶标包括半胱氨酸蛋白酶3CLpro(也称为Mpro)和PLpro,它们对于产生新的成熟病毒粒子至关重要[7,8]。另一个潜在的靶标是RdRP,它是病毒基因组复制所必需的。为了将新的药物疗法快速应用于临床,药物再利用已经得到了广泛的研究。这样做的好处是,潜在的治疗方法已经被批准用于人类。虚拟筛查已被广泛用于帮助重新利用现有的COVID-19药物治疗[2,10]。这种方法比传统的基于实验室的测试更快速、更经济。许多潜在的药物治疗已被确定使用这一过程。这些药物包括以前用于治疗埃博拉病毒和丙型肝炎病毒的瑞德西韦,以及以前用于治疗呼吸道合胞病毒感染、丙型肝炎和某些出血热的利巴韦林。据报道,Remdesivir靶向RdRP,而利巴韦林同时靶向3CLpro和RdRP[2,6,11]。最近,美国FDA (MD, USA)批准了瑞德西韦用于12岁以上的COVID-19患者。然而,世界卫生组织(瑞士日内瓦)发布了一项未发表的临床试验数据,表明瑞德西韦不能降低COVID-19重症患者的死亡率、缩短住院时间或减少对通气的需求。其他药物组合的临床试验仍在进行中。开发新的治疗性抗病毒药物显然需要开发新的靶向抗病毒药物来治疗SARS-CoV-2感染。然而,药物发现和开发过程可能过于漫长,无法应对当前的大流行。对任何针对SARS-CoV-2靶点的潜在新疗法的结构-活性关系的基本理解是至关重要的。冠状病毒之间氨基酸序列同源性高。例如,SARS-CoV-2刺突糖蛋白和3CLpro与SARS-CoV的氨基酸序列同源性分别为76%和96%[2,10]。在序列同源性高且蛋白质结构相似的情况下,针对SARS-CoV开发的抑制剂可能对SARS-CoV-2有效[5,9]。如果没有,这些化合物至少可能为优化和随后的药物开发提供良好的线索[5,9]。计算建模和虚拟筛选可以帮助快速识别导联,在实验室功能测试和x射线晶体学的支持下,生成新的结构-活性关系,进一步优化导联。当设计检测潜在治疗方法时,重要的是要考虑目标蛋白的功能状态。例如,来自sars冠状病毒的3CLpro被发现仅在溶液[13]中作为二聚体具有活性。研究还表明,在C端或n端附加的亲和力标签可能会降低其酶活性[13]。由于与SARS-CoV-2的3CLpro序列高度同源,在建立合适的筛选方法时应检查这些标准[2,13]。基于fret的荧光底物试验和未标记的3CLpro已被用于确定靶向SARS-CoV 3CLpro的潜在药物样配体的IC50[8,13]。同样,等温滴定量热法(ITC)已被用于确定潜在药物[8]的结合亲和力。宿主编码的治疗靶点ACE2在肺泡上皮细胞(以及心脏、肾脏和睾丸)上表达,是SARS-Cov-2刺突糖蛋白结合[3]的靶点。 这种相互作用允许SARS-CoV-2膜与宿主细胞融合并进入病毒基因组[3]。已经试验了一种可能的治疗方法,使用重组可溶形式的人ACE2来阻断Spike与细胞表面表达的ACE2的结合。这可以阻止病毒进入宿主细胞,阻断病毒复制,减少病毒负担[3,14]。我们使用无标记表面等离子体共振(SPR)技术分析ACE2和Spike糖蛋白之间的生物分子相互作用的未发表的观察结果很好地证明了这一点。与可溶性ACE2预孵育的穗糖蛋白显著降低了固定在SPR传感器表面的穗与ACE2的结合。抗炎药物COVID-19的治疗不仅限于抗病毒药物。它们还包括使用药物来减少宿主的免疫反应。一个有文献记载的问题是Ang II的增加,除了其心血管作用外,它还作为炎症蛋白[16]。angii被ace2[16]降解。刺突-ACE2复合物的内吞作用,即SARS-CoV-2进入细胞的机制,被认为会导致ACE2可用性降低,进而导致Ang II的增加。高水平的Ang II结合AT1导致信号级联,导致促炎反应[16]。这反过来又可能导致急性呼吸窘迫。有人提出AT1阻滞剂可用于与Ang II竞争AT1结合位点[16]。对各种AT1阻滞剂的亲和力已经用放射性配体结合试验[16]来测量,[16]是药物发现过程中另一个有用的工具。其他减少炎症反应的方法包括使用地塞米松等皮质类固醇治疗,使用呼吸机的患者死亡率可降低三分之一。这种治疗方法对严重感染的患者很有用,可以减少副作用。然而,皮质类固醇没有抗病毒特性,不推荐用于早期疾病bbb的治疗。以抗体为基础的疗法输注来自covid -19感染后康复的献血者的血浆,其中含有针对SARS-CoV-2抗原的抗体,已被预防性地用于预防高危人群的感染[18,19]。然而,来自单个感染后供体的血浆最多只能满足三个受体。使用靶向病毒刺突糖蛋白的重组单克隆抗体并大量生产,克服了感染后血浆供应有限的问题[18,19]。单克隆抗体在重症患者中的临床试验正在进行中,但有效的治疗方法尚未被批准[1,18]。所有这些蛋白质输注疗法(抗体和可溶性ACE2)都受到宿主蛋白质降解的限制,需要定期再治疗以维持保护。与大多数病毒性疾病一样,通过全球疫苗接种计划预防在宿主中建立感染和随后的病毒传播被认为是最成功的方法。在等待有效疫苗获得批准的同时,许多国家正在实施社交距离和使用面罩来控制病毒bbb的传播。对有效疫苗的需求是显而易见的。据报道,目前正在进行150多项疫苗试验[1,4]。大多数疫苗针对SARS-CoV-2刺突糖蛋白[4]。各种类型的疫苗目前正在进行不同阶段的临床试验,包括蛋白质亚单位疫苗、基于rna的疫苗和复制病毒载体疫苗[4]。辉瑞(美国纽约州)和BioNTech(德国美因茨)最近宣布,他们的密码子优化mRNA疫苗(BNT162b1)三期试验在预防COVID-19感染方面的有效性为90%。然而,这些数据还没有经过同行评审。一种有效的疫苗将有助于防止疾病的传播,使社会保持距离的措施得以缓解,并使社会恢复正常。结论:虽然药物再利用取得了一些有限的成功,但很明显,需要一种靶向抗病毒药物来治疗持续的SARS-CoV-2感染患者。针对SARS-CoV-2的疫苗最有可能防止感染在宿主体内建立,从而防止病毒的进一步传播,从而降低全球死亡率。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
审稿时长
14 weeks
期刊最新文献
Widened scope of drug repurposing/chiral switches, elements of secondary pharmaceuticals: the quinine/quinidine case Codeine dysregulates ribosome biogenesis in Escherichia coli with DNA double-strand breaks to chart path to new classes of antibiotics PepFun 2.0: improved protocols for the analysis of natural and modified peptides Development of a novel chemoinformatic tool for natural product databases Computational approaches to targeting protein–protein interactions in cancer: a pathway to drug discovery
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1