首页 > 最新文献

Frontiers in drug discovery最新文献

英文 中文
The critical role of mode of action studies in kinetoplastid drug discovery. 作用模式研究在发现新药中的关键作用。
Pub Date : 2023-05-10 DOI: 10.3389/fddsv.2023.1185679
Alan H Fairlamb, Susan Wyllie

Understanding the target and mode of action of compounds identified by phenotypic screening can greatly facilitate the process of drug discovery and development. Here, we outline the tools currently available for target identification against the neglected tropical diseases, human African trypanosomiasis, visceral leishmaniasis and Chagas' disease. We provide examples how these tools can be used to identify and triage undesirable mechanisms, to identify potential toxic liabilities in patients and to manage a balanced portfolio of target-based campaigns. We review the primary targets of drugs that are currently in clinical development that were initially identified via phenotypic screening, and whose modes of action affect protein turnover, RNA trans-splicing or signalling in these protozoan parasites.

了解通过表型筛选确定的化合物的作用靶点和作用模式可极大地促进药物发现和开发过程。在此,我们概述了目前针对被忽视的热带疾病、非洲锥虫病、内脏利什曼病和南美锥虫病的靶点鉴定工具。我们将举例说明如何利用这些工具识别和分流不良机制、识别患者的潜在毒性责任以及管理基于靶点的平衡组合。我们回顾了目前处于临床开发阶段的药物的主要靶点,这些靶点最初是通过表型筛选确定的,其作用模式会影响这些原生动物寄生虫体内的蛋白质周转、RNA 转拼接或信号传导。
{"title":"The critical role of mode of action studies in kinetoplastid drug discovery.","authors":"Alan H Fairlamb, Susan Wyllie","doi":"10.3389/fddsv.2023.1185679","DOIUrl":"10.3389/fddsv.2023.1185679","url":null,"abstract":"<p><p>Understanding the target and mode of action of compounds identified by phenotypic screening can greatly facilitate the process of drug discovery and development. Here, we outline the tools currently available for target identification against the neglected tropical diseases, human African trypanosomiasis, visceral leishmaniasis and Chagas' disease. We provide examples how these tools can be used to identify and triage undesirable mechanisms, to identify potential toxic liabilities in patients and to manage a balanced portfolio of target-based campaigns. We review the primary targets of drugs that are currently in clinical development that were initially identified via phenotypic screening, and whose modes of action affect protein turnover, RNA trans-splicing or signalling in these protozoan parasites.</p>","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7614965/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10173670","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Generation of a potent anti-PD-L1-CD47 bispecific antibody with a strong therapeutic and safety profile for cancer immunotherapy 产生一种有效的抗pd - l1 - cd47双特异性抗体,具有很强的治疗性和安全性,可用于癌症免疫治疗
Pub Date : 2023-05-09 DOI: 10.3389/fddsv.2023.1182146
Irene Tang, L. Schwimmer, Shenda Gu, Wei Wei Prior, Hieu Van Tran, Allan Chan, Anna McClain, C. Fraser, Chunyang Sun, M. Si, Guijiang Wang, Yunxia Zhao, Ning Zhang, Jiayu Fu, Mengxin Liu, Chuanzeng Cao, Shihao Chen
Cell surface molecules PD-L1 and CD47 are potent inhibitors of adaptive and innate anti-cancer immunity. We sought to generate a safe, therapeutic, bispecific antibody specifically targeting, and blocking both PD-L1 and CD47 inhibitory activity. Novel anti-PDL-1 and anti-CD47 antibodies with favorable inhibitory activity, were humanized and constructed into a unique bi-specific antibody intended for clinical use. Previous pre-clinical and clinical studies using anti-CD47 antibodies indicated anemia and thrombocytopenia as potential risks. QL401 is a PD-L1 x CD47 bispecific antibody engineered to reduce effect on red blood cells while retaining potent phagocytic activation of macrophages in vitro and delayed tumor growth in vivo. QL401 comprises three functional components: a PD-L1 binding Fab arm, a CD47 binding scFv arm, and a human IgG4 backbone. The PD-L1 binding arm provides both tumor targeting and blocking of PD-1 for reactivating T cells. The CD47 arm blocks the binding of SIRPα, while the IgG4 Fc retains Fc gamma receptor binding to provide a phagocytic signal. In preclinical efficacy studies, QL401 potently blocked SIRPα to promote phagocytosis of tumor cells with sub-nanomolar potency. In vivo efficacy studies in mouse xenograft tumor models showed QL401 to be comparable or superior to PD-L1 or CD47 monoclonal antibodies alone or in combination. In vitro safety evaluation of QL401 showed significantly reduced binding and phagocytosis of red blood cells, in contrast to CD47 monoclonal antibodies. In addition, QL401 did not induce hemagglutination. In non-human primates, QL401 was well tolerated up to 100 mg/kg without reduction of red blood cells or platelets below the normal range. QL401 is presently in a human phase I safety study.
细胞表面分子PD-L1和CD47是适应性和先天抗癌免疫的有效抑制剂。我们寻求产生一种安全、治疗性、双特异性的抗体,特异性靶向并阻断PD-L1和CD47的抑制活性。将具有良好抑制活性的新型抗pdl -1和抗cd47抗体人源化并构建成一种独特的双特异性抗体,用于临床应用。先前使用抗cd47抗体的临床前和临床研究表明,贫血和血小板减少是潜在的风险。QL401是一种PD-L1 x CD47双特异性抗体,旨在减少对红细胞的影响,同时在体外保持巨噬细胞的有效吞噬激活,并在体内延缓肿瘤生长。QL401由三个功能组件组成:PD-L1结合Fab臂、CD47结合scFv臂和人IgG4骨干。PD-L1结合臂提供肿瘤靶向和阻断PD-1来重新激活T细胞。CD47臂阻断SIRPα的结合,而IgG4 Fc保留Fc γ受体结合以提供吞噬信号。在临床前疗效研究中,QL401有效阻断SIRPα,以亚纳摩尔的效力促进肿瘤细胞的吞噬。在小鼠异种移植肿瘤模型中的体内疗效研究表明,QL401与单独或联合使用的PD-L1或CD47单克隆抗体相当或优于单克隆抗体。体外安全性评价显示,与CD47单克隆抗体相比,QL401对红细胞的结合和吞噬作用显著降低。此外,QL401不诱导血凝。在非人类灵长类动物中,QL401耐受性良好,高达100 mg/kg,红细胞或血小板未减少到正常范围以下。QL401目前正在进行人体I期安全性研究。
{"title":"Generation of a potent anti-PD-L1-CD47 bispecific antibody with a strong therapeutic and safety profile for cancer immunotherapy","authors":"Irene Tang, L. Schwimmer, Shenda Gu, Wei Wei Prior, Hieu Van Tran, Allan Chan, Anna McClain, C. Fraser, Chunyang Sun, M. Si, Guijiang Wang, Yunxia Zhao, Ning Zhang, Jiayu Fu, Mengxin Liu, Chuanzeng Cao, Shihao Chen","doi":"10.3389/fddsv.2023.1182146","DOIUrl":"https://doi.org/10.3389/fddsv.2023.1182146","url":null,"abstract":"Cell surface molecules PD-L1 and CD47 are potent inhibitors of adaptive and innate anti-cancer immunity. We sought to generate a safe, therapeutic, bispecific antibody specifically targeting, and blocking both PD-L1 and CD47 inhibitory activity. Novel anti-PDL-1 and anti-CD47 antibodies with favorable inhibitory activity, were humanized and constructed into a unique bi-specific antibody intended for clinical use. Previous pre-clinical and clinical studies using anti-CD47 antibodies indicated anemia and thrombocytopenia as potential risks. QL401 is a PD-L1 x CD47 bispecific antibody engineered to reduce effect on red blood cells while retaining potent phagocytic activation of macrophages in vitro and delayed tumor growth in vivo. QL401 comprises three functional components: a PD-L1 binding Fab arm, a CD47 binding scFv arm, and a human IgG4 backbone. The PD-L1 binding arm provides both tumor targeting and blocking of PD-1 for reactivating T cells. The CD47 arm blocks the binding of SIRPα, while the IgG4 Fc retains Fc gamma receptor binding to provide a phagocytic signal. In preclinical efficacy studies, QL401 potently blocked SIRPα to promote phagocytosis of tumor cells with sub-nanomolar potency. In vivo efficacy studies in mouse xenograft tumor models showed QL401 to be comparable or superior to PD-L1 or CD47 monoclonal antibodies alone or in combination. In vitro safety evaluation of QL401 showed significantly reduced binding and phagocytosis of red blood cells, in contrast to CD47 monoclonal antibodies. In addition, QL401 did not induce hemagglutination. In non-human primates, QL401 was well tolerated up to 100 mg/kg without reduction of red blood cells or platelets below the normal range. QL401 is presently in a human phase I safety study.","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"91355037","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Editorial: Development/repurposing of drugs to tackle the multiple variants of SARS-CoV-2 社论:开发/重新调整药物用途以应对严重急性呼吸系统综合征冠状病毒2型的多种变体
Pub Date : 2023-02-27 DOI: 10.3389/fddsv.2023.1157688
D. Gambino
COVID-19, the severe acute respiratory syndrome caused by Coronavirus (SARS-CoV-2) and identified for the first time in China in 2019, was recognized in 2020 as a global pandemic by the World Health Organization (Wu et al., 2020; WHO, 2023). Although elder people and all those with underlying medical conditions like cardiovascular disease, diabetes, chronic respiratory disease, or cancer are more likely to develop serious illness, people at any age can become seriously ill or die (WHO, 2023). The efforts of pharmaceutical companies and academia have successfully led to several vaccines against this virus in an unprecedented short period of time. Although vaccines provide protection to healthy people, they could be not effective for immune compromised individuals or those bearing some risky pathological comorbidities. Additionally, mutations could generate viral variants unaffected by currently available vaccines. Therefore, new chemotherapeutic agents are urgently needed for the treatment of SARS-CoV-2 in order to reduce virus dissemination and mortality. Although huge efforts are beingmade since 2020 towards the development of new drugs or the repurposing of already approved drugs to other targets, which would lead to a significant drop in the approval time of these drugs, drugs for the treatment of COVID-19 are not yet a reality (Ashburn and Thor, 2004; Nosengo, 2016; WHO, 2023). At present, there is a clinical need for direct-acting antivirals targeting SARS-CoV-2 to complement existing therapeutic strategies. Accordingly, the aim of this Research Topic of Frontiers in Drug Discovery, Antiinfective Agents, is to collect latest research on the topic focused on:
新冠肺炎是由冠状病毒(SARS-CoV-2)引起的严重急性呼吸综合征,于2019年在中国首次被发现,2020年被世界卫生组织确认为全球大流行(Wu et al.,2020;世界卫生组织,2023)。尽管老年人和所有患有心血管疾病、糖尿病、慢性呼吸道疾病或癌症等潜在疾病的人更有可能患上严重疾病,但任何年龄的人都可能患上重症或死亡(世界卫生组织,2023)。制药公司和学术界的努力在前所未有的短时间内成功研制出了几种针对这种病毒的疫苗。尽管疫苗为健康人提供了保护,但对免疫受损的个体或患有一些危险病理合并症的人可能无效。此外,突变可能产生不受当前可用疫苗影响的病毒变体。因此,迫切需要新的化疗药物来治疗严重急性呼吸系统综合征冠状病毒2型,以减少病毒传播和死亡率。尽管自2020年以来,正在为开发新药或将已批准的药物重新用于其他目标做出巨大努力,这将导致这些药物的批准时间大幅缩短,但治疗新冠肺炎的药物尚未成为现实(Ashburn和Thor,2004;诺森戈,2016;世界卫生组织,2023)。目前,临床上需要针对严重急性呼吸系统综合征冠状病毒2型的直接作用抗病毒药物来补充现有的治疗策略。因此,本研究主题“药物发现的前沿,抗感染剂”的目的是收集关于以下主题的最新研究:
{"title":"Editorial: Development/repurposing of drugs to tackle the multiple variants of SARS-CoV-2","authors":"D. Gambino","doi":"10.3389/fddsv.2023.1157688","DOIUrl":"https://doi.org/10.3389/fddsv.2023.1157688","url":null,"abstract":"COVID-19, the severe acute respiratory syndrome caused by Coronavirus (SARS-CoV-2) and identified for the first time in China in 2019, was recognized in 2020 as a global pandemic by the World Health Organization (Wu et al., 2020; WHO, 2023). Although elder people and all those with underlying medical conditions like cardiovascular disease, diabetes, chronic respiratory disease, or cancer are more likely to develop serious illness, people at any age can become seriously ill or die (WHO, 2023). The efforts of pharmaceutical companies and academia have successfully led to several vaccines against this virus in an unprecedented short period of time. Although vaccines provide protection to healthy people, they could be not effective for immune compromised individuals or those bearing some risky pathological comorbidities. Additionally, mutations could generate viral variants unaffected by currently available vaccines. Therefore, new chemotherapeutic agents are urgently needed for the treatment of SARS-CoV-2 in order to reduce virus dissemination and mortality. Although huge efforts are beingmade since 2020 towards the development of new drugs or the repurposing of already approved drugs to other targets, which would lead to a significant drop in the approval time of these drugs, drugs for the treatment of COVID-19 are not yet a reality (Ashburn and Thor, 2004; Nosengo, 2016; WHO, 2023). At present, there is a clinical need for direct-acting antivirals targeting SARS-CoV-2 to complement existing therapeutic strategies. Accordingly, the aim of this Research Topic of Frontiers in Drug Discovery, Antiinfective Agents, is to collect latest research on the topic focused on:","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45053551","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Numaswitch, a biochemical platform for the efficient production of disulfide-rich pepteins Numaswitch,一个高效生产富含二硫化物的肽的生化平台
Pub Date : 2023-02-21 DOI: 10.3389/fddsv.2023.1082058
Bach-Ngan Nguyen, Florian Tieves, Florian G. Neusius, H. Götzke, L. Schmitt, C. Schwarz
The application of long-chained peptides (+30 aa) and relatively short proteins (<300 aa) has experienced an increasing interest in recent years. However, a reliable production platform is still missing since manufacturing is challenged by inherent problems such as mis-folding, aggregation, and low production yields. And neither chemical synthesis nor available recombinant approaches are effective and efficient. This in particular holds true for disulfide-rich targets where the correct isomer needs to be formed. With the technology Numaswitch, we have now developed a biochemical tool that circumvents existing limitations and serves as first production platform for pepteins, hard-to-be-produced peptides and proteins between 30 and 300 amino acids in length, including disulfide-rich candidates. Numaswitch is based on bifunctional Switchtag proteins that force the high-titer expression of pure inclusion bodies and simultaneously assist in the efficient refolding of pepteins into functional pepteins. Here, we demonstrate the successful application of the Numaswitch platform for disulfide-containing pepteins, such as an antimicrobial fusion peptide, a single-chain variable fragment (scFv), a camelid heavy chain antibody fragment (VHH) and the human epidermal growth factor.
近年来,人们对长链肽(+30个氨基酸)和相对短的蛋白质(<300个氨基酸)的应用越来越感兴趣。然而,由于制造业受到固有问题的挑战,如错误折叠、聚集和低产量,因此仍然缺乏可靠的生产平台。化学合成和现有的重组方法都不是有效的。这尤其适用于需要形成正确异构体的富含二硫化物的靶标。利用Numaswitch技术,我们现在开发了一种生化工具,该工具绕过了现有的限制,并作为蛋白质蛋白的第一个生产平台,蛋白质蛋白是一种难以生产的肽和蛋白质,长度在30至300个氨基酸之间,包括富含二硫化物的候选者。Numaswitch基于双功能Switchtag蛋白,该蛋白迫使纯包涵体的高滴度表达,同时帮助蛋白质蛋白有效地重折叠为功能性蛋白质蛋白。在这里,我们展示了Numaswitch平台在含二硫化物的蛋白肽中的成功应用,如抗微生物融合肽、单链可变片段(scFv)、骆驼重链抗体片段(VHH)和人表皮生长因子。
{"title":"Numaswitch, a biochemical platform for the efficient production of disulfide-rich pepteins","authors":"Bach-Ngan Nguyen, Florian Tieves, Florian G. Neusius, H. Götzke, L. Schmitt, C. Schwarz","doi":"10.3389/fddsv.2023.1082058","DOIUrl":"https://doi.org/10.3389/fddsv.2023.1082058","url":null,"abstract":"The application of long-chained peptides (+30 aa) and relatively short proteins (<300 aa) has experienced an increasing interest in recent years. However, a reliable production platform is still missing since manufacturing is challenged by inherent problems such as mis-folding, aggregation, and low production yields. And neither chemical synthesis nor available recombinant approaches are effective and efficient. This in particular holds true for disulfide-rich targets where the correct isomer needs to be formed. With the technology Numaswitch, we have now developed a biochemical tool that circumvents existing limitations and serves as first production platform for pepteins, hard-to-be-produced peptides and proteins between 30 and 300 amino acids in length, including disulfide-rich candidates. Numaswitch is based on bifunctional Switchtag proteins that force the high-titer expression of pure inclusion bodies and simultaneously assist in the efficient refolding of pepteins into functional pepteins. Here, we demonstrate the successful application of the Numaswitch platform for disulfide-containing pepteins, such as an antimicrobial fusion peptide, a single-chain variable fragment (scFv), a camelid heavy chain antibody fragment (VHH) and the human epidermal growth factor.","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47786351","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
HMM-based profiling identifies the binding to divalent cations and nucleotides as common denominators of suramin targets 基于hmm的分析识别结合二价阳离子和核苷酸作为苏拉明目标的共同分母
Pub Date : 2023-02-10 DOI: 10.3389/fddsv.2023.1112992
Dennis A. Hauser, P. Mäser
Introduction: Suramin is one of the pharmacopeia’s most promiscuous drugs. Originally developed for African trypanosomiasis, suramin was also used for onchocerciasis and it has been proposed as an anticancer agent, antiviral drug, therapy for arthritis, autism, and antidote for snake bites. Target proteins of suramin have been described from different species. Here we identify the common motifs among these various targets, aiming to explain the promiscuous nature of suramin. Methods: We have searched for suramin target proteins in the literature and in chemical databases. Applying rigorous inclusion criteria, a list of 44 diverse proteins was assembled with experimental evidence for direct interaction with, and inhibition by, suramin. Hidden Markov model-based target profiling was performed by running the full set of Pfam protein family domains against these proteins. Results: Common denominators were identified by mapping the identified Pfam domains to molecular function gene ontology terms. This in silico pipeline identified nucleotide binding, nucleic acid binding, and binding to divalent cations as the most common denominators of the suramin targets. Discussion: Our results suggest that the extraordinary polypharmacology of suramin may be caused by its ability to inhibit the interaction of proteins with nucleotides or nucleic acids and with divalent cations (Mg2+, Ca2+, Zn2+). Suramin is well known to inhibit nucleotide receptors and nucleic acid-binding enzymes. The association with divalent cations is new and might be key towards the design of better, more selective inhibitors.
苏拉明是药典中最混杂的药物之一。苏拉明最初是为非洲锥虫病开发的,也被用于盘尾丝虫病,它被认为是一种抗癌剂、抗病毒药物、治疗关节炎、自闭症和蛇咬伤的解药。苏拉明的靶蛋白已从不同物种中进行了描述。在这里,我们确定了这些不同靶标之间的共同主题,旨在解释苏拉明的混杂性质。方法:我们在文献和化学数据库中搜索苏拉明靶蛋白。应用严格的纳入标准,列出了44种不同的蛋白质,并提供了与苏拉明直接相互作用和抑制苏拉明的实验证据。通过针对这些蛋白质运行全套Pfam蛋白质家族结构域来进行基于隐马尔可夫模型的靶标图谱分析。结果:通过将已识别的Pfam结构域映射到分子功能基因本体论术语来识别共分母。这一计算机管道将核苷酸结合、核酸结合和与二价阳离子的结合确定为苏拉明靶标最常见的分母。讨论:我们的研究结果表明,苏拉明非凡的多药性可能是由其抑制蛋白质与核苷酸或核酸以及与二价阳离子(Mg2+、Ca2+、Zn2+)相互作用的能力引起的。众所周知,苏拉明能抑制核苷酸受体和核酸结合酶。与二价阳离子的结合是新的,可能是设计更好、更具选择性的抑制剂的关键。
{"title":"HMM-based profiling identifies the binding to divalent cations and nucleotides as common denominators of suramin targets","authors":"Dennis A. Hauser, P. Mäser","doi":"10.3389/fddsv.2023.1112992","DOIUrl":"https://doi.org/10.3389/fddsv.2023.1112992","url":null,"abstract":"Introduction: Suramin is one of the pharmacopeia’s most promiscuous drugs. Originally developed for African trypanosomiasis, suramin was also used for onchocerciasis and it has been proposed as an anticancer agent, antiviral drug, therapy for arthritis, autism, and antidote for snake bites. Target proteins of suramin have been described from different species. Here we identify the common motifs among these various targets, aiming to explain the promiscuous nature of suramin. Methods: We have searched for suramin target proteins in the literature and in chemical databases. Applying rigorous inclusion criteria, a list of 44 diverse proteins was assembled with experimental evidence for direct interaction with, and inhibition by, suramin. Hidden Markov model-based target profiling was performed by running the full set of Pfam protein family domains against these proteins. Results: Common denominators were identified by mapping the identified Pfam domains to molecular function gene ontology terms. This in silico pipeline identified nucleotide binding, nucleic acid binding, and binding to divalent cations as the most common denominators of the suramin targets. Discussion: Our results suggest that the extraordinary polypharmacology of suramin may be caused by its ability to inhibit the interaction of proteins with nucleotides or nucleic acids and with divalent cations (Mg2+, Ca2+, Zn2+). Suramin is well known to inhibit nucleotide receptors and nucleic acid-binding enzymes. The association with divalent cations is new and might be key towards the design of better, more selective inhibitors.","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46032731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In silico identification of potential inhibitors of acyl carrier protein reductase and acetyl CoA carboxylase of Plasmodium falciparum in antimalarial therapy 恶性疟原虫酰基载体蛋白还原酶和乙酰辅酶a羧化酶潜在抑制剂在抗疟治疗中的应用
Pub Date : 2023-01-20 DOI: 10.3389/fddsv.2023.1087008
Elliasu Y. Salifu, James Abugri, Issahaku A Rashid, F. Osei, Joseph Atia Ayariga
Malaria caused by Plasmodium falciparum, remains one of the most fatal parasitic diseases that has affected nearly a third of the world’s population. The major impediment to the treatment of malaria is the emergence of resistance of the P. falciparum parasite to current anti-malaria therapeutics such as Artemisinin (ART)-based combination therapy (ACT). This has resulted in countless efforts to develop novel therapeutics that will counter this resistance with the aim to control and eradicate the disease. The application of in silico modelling techniques has gained a lot of recognition in antimalarial research in recent times through the identification of biological components of the parasite for rational drug design. In this study we employed various in silico techniques such as the Virtual screening, molecular docking and molecular dynamic simulations to identify potential new inhibitors of biotin acetyl-coenzyme A (CoA) carboxylase and enoyl-acyl carrier reductase, two enzyme targets that play a crucial role in fatty acid synthesis in the Plasmodium parasite. Initially, nine hit compounds were identified for each of the two enzymes from the ZINCPharmer database. Subsequently, all hit compounds bind favourably to the active sites of the two enzymes as well as show excellent pharmacokinetic properties. Three 3) of the hits for the biotin acetyl-coenzyme A (CoA) carboxylase and six 6) of the enoyl-acyl carrier reductase showed good toxicity properties. The compounds were further evaluated based on the Molecular Dynamics simulation that confirmed the binding stability of the compounds to the targeted proteins. Overall, the lead compounds ZINC38980461, ZINC05378039, and ZINC15772056, were identified for acetyl-coenzyme A (CoA) carboxylase whiles ZINC94085628, ZINC93656835, ZINC94080670, ZINC1774609, ZINC94821232 and ZINC94919772 were identified as lead compounds for enoyl-acyl carrier reductase. The identified compounds can be developed as a treatment option for the malaria disease although, experimental validation is suggested for further evaluation of the work.
恶性疟原虫引起的疟疾仍然是最致命的寄生虫病之一,影响了世界上近三分之一的人口。疟疾治疗的主要障碍是恶性疟原虫对目前的抗疟疾疗法产生耐药性,如基于青蒿素(ART)的联合疗法(ACT)。这导致了无数人努力开发新的治疗方法,以对抗这种耐药性,从而控制和根除这种疾病。近年来,通过鉴定寄生虫的生物成分以进行合理的药物设计,计算机建模技术在抗疟研究中的应用得到了很多认可。在这项研究中,我们采用了各种计算机技术,如虚拟筛选、分子对接和分子动力学模拟,以确定生物素乙酰辅酶A羧化酶和烯酰基载体还原酶的潜在新抑制剂,这两个酶靶点在疟原虫的脂肪酸合成中起着至关重要的作用。最初,从ZINCPharmer数据库中,两种酶中的每一种都鉴定出了9种命中化合物。随后,所有命中的化合物都与两种酶的活性位点有利地结合,并显示出优异的药代动力学特性。生物素乙酰辅酶A(CoA)羧化酶的三个3)命中率和烯酰基载体还原酶的六个6)命中率显示出良好的毒性特性。基于分子动力学模拟进一步评估化合物,该模拟证实了化合物与靶蛋白的结合稳定性。总的来说,乙酰辅酶A(CoA)羧化酶的先导化合物锌38980461、锌05378039和锌15772056被鉴定,而烯酰基载体还原酶的先导化合物为锌94085628、锌9356835、锌94080670、锌1774609、锌94821232和锌94919772。已鉴定的化合物可以作为疟疾的治疗选择,尽管建议进行实验验证以进一步评估这项工作。
{"title":"In silico identification of potential inhibitors of acyl carrier protein reductase and acetyl CoA carboxylase of Plasmodium falciparum in antimalarial therapy","authors":"Elliasu Y. Salifu, James Abugri, Issahaku A Rashid, F. Osei, Joseph Atia Ayariga","doi":"10.3389/fddsv.2023.1087008","DOIUrl":"https://doi.org/10.3389/fddsv.2023.1087008","url":null,"abstract":"Malaria caused by Plasmodium falciparum, remains one of the most fatal parasitic diseases that has affected nearly a third of the world’s population. The major impediment to the treatment of malaria is the emergence of resistance of the P. falciparum parasite to current anti-malaria therapeutics such as Artemisinin (ART)-based combination therapy (ACT). This has resulted in countless efforts to develop novel therapeutics that will counter this resistance with the aim to control and eradicate the disease. The application of in silico modelling techniques has gained a lot of recognition in antimalarial research in recent times through the identification of biological components of the parasite for rational drug design. In this study we employed various in silico techniques such as the Virtual screening, molecular docking and molecular dynamic simulations to identify potential new inhibitors of biotin acetyl-coenzyme A (CoA) carboxylase and enoyl-acyl carrier reductase, two enzyme targets that play a crucial role in fatty acid synthesis in the Plasmodium parasite. Initially, nine hit compounds were identified for each of the two enzymes from the ZINCPharmer database. Subsequently, all hit compounds bind favourably to the active sites of the two enzymes as well as show excellent pharmacokinetic properties. Three 3) of the hits for the biotin acetyl-coenzyme A (CoA) carboxylase and six 6) of the enoyl-acyl carrier reductase showed good toxicity properties. The compounds were further evaluated based on the Molecular Dynamics simulation that confirmed the binding stability of the compounds to the targeted proteins. Overall, the lead compounds ZINC38980461, ZINC05378039, and ZINC15772056, were identified for acetyl-coenzyme A (CoA) carboxylase whiles ZINC94085628, ZINC93656835, ZINC94080670, ZINC1774609, ZINC94821232 and ZINC94919772 were identified as lead compounds for enoyl-acyl carrier reductase. The identified compounds can be developed as a treatment option for the malaria disease although, experimental validation is suggested for further evaluation of the work.","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43816255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 4
The use of cyclic peptide antigens to generate LRP8 specific antibodies 利用环肽抗原产生LRP8特异性抗体
Pub Date : 2023-01-12 DOI: 10.3389/fddsv.2022.1093153
M. Argiriadi, Kangwen Deng, D. Egan, Lei Gao, F. Gizatullin, J. Harlan, Denise Karaoglu Hanzatian, W. Qiu, Ruth Villanueva, Andrew Goodearl
LRP8 is a member of the LDLR-like protein family. It is a transport receptor, which can be used in the design of antibodies specific for investigating increasing exposure to therapeutics with respect to the blood brain barrier (BBB). In this study, a LRP8 peptide immunization strategy was implemented to generate antibodies to a specific epitope of the CR1 domain of LRP8 that could enable transport function and cross-react in mice, cynomolgus monkeys and humans. Additionally, a cyclized peptide immunogen was designed to conserve the structural β-hairpin element observed in a previously solved crystal structure of a related CR domain. As a result of this structure-based antigenic design, an LRP8 specific antibody, 11H1, was selected and characterized in ligand binding assays and crystallographic structure determination. The high-resolution structure of the 11H1 Fab complexed to the cyclized CR1 peptide revealed key interactions driving epitope recognition that were confirmed using a site-directed mutagenesis approach. A critical observation was that the identified structural CR1 epitope of 11H1 did not compete with reelin’s recognition of CR1 allowing for simultaneous binding. This was predicted by an in silico ternary model and confirmed by reelin binding data. These simultaneous binding events (11H1/CR1/reelin) could therefore enable the CR1 domain of LRP8, 11H1 and reelin to be used as a “BBB transporter” ternary complex in the design of therapeutic proteins. More importantly, 11H1 showed enhanced brain penetration after systemic intravenous dosing in a mouse study, which confirmed its potential function as BBB transporter for therapeutic proteins.
LRP8是LDLR样蛋白家族的成员。它是一种转运受体,可用于设计特异性抗体,以研究血脑屏障(BBB)治疗方法暴露量的增加。在本研究中,实施了LRP8肽免疫策略,以产生针对LRP8 CR1结构域特定表位的抗体,该抗体可以在小鼠、食蟹猴和人类中实现转运功能和交叉反应。此外,设计了一种环化肽免疫原,以保存在相关CR结构域的先前解决的晶体结构中观察到的结构β-发夹元件。作为这种基于结构的抗原设计的结果,选择了LRP8特异性抗体11H1,并在配体结合测定和晶体结构测定中进行了表征。与环化CR1肽复合的11H1Fab的高分辨率结构揭示了驱动表位识别的关键相互作用,这些相互作用使用定点诱变方法得到了证实。一个关键的观察结果是,11H1的已鉴定的结构CR1表位与reelin对CR1的识别不竞争,从而允许同时结合。这是由硅内三元模型预测的,并由reelin结合数据证实。因此,这些同时发生的结合事件(11H1/CR1/reelin)可以使LRP8、11H1和reelin的CR1结构域在治疗蛋白的设计中用作“血脑屏障转运蛋白”三元复合物。更重要的是,在一项小鼠研究中,11H1在全身静脉给药后显示出增强的脑渗透性,这证实了其作为治疗蛋白的血脑屏障转运蛋白的潜在功能。
{"title":"The use of cyclic peptide antigens to generate LRP8 specific antibodies","authors":"M. Argiriadi, Kangwen Deng, D. Egan, Lei Gao, F. Gizatullin, J. Harlan, Denise Karaoglu Hanzatian, W. Qiu, Ruth Villanueva, Andrew Goodearl","doi":"10.3389/fddsv.2022.1093153","DOIUrl":"https://doi.org/10.3389/fddsv.2022.1093153","url":null,"abstract":"LRP8 is a member of the LDLR-like protein family. It is a transport receptor, which can be used in the design of antibodies specific for investigating increasing exposure to therapeutics with respect to the blood brain barrier (BBB). In this study, a LRP8 peptide immunization strategy was implemented to generate antibodies to a specific epitope of the CR1 domain of LRP8 that could enable transport function and cross-react in mice, cynomolgus monkeys and humans. Additionally, a cyclized peptide immunogen was designed to conserve the structural β-hairpin element observed in a previously solved crystal structure of a related CR domain. As a result of this structure-based antigenic design, an LRP8 specific antibody, 11H1, was selected and characterized in ligand binding assays and crystallographic structure determination. The high-resolution structure of the 11H1 Fab complexed to the cyclized CR1 peptide revealed key interactions driving epitope recognition that were confirmed using a site-directed mutagenesis approach. A critical observation was that the identified structural CR1 epitope of 11H1 did not compete with reelin’s recognition of CR1 allowing for simultaneous binding. This was predicted by an in silico ternary model and confirmed by reelin binding data. These simultaneous binding events (11H1/CR1/reelin) could therefore enable the CR1 domain of LRP8, 11H1 and reelin to be used as a “BBB transporter” ternary complex in the design of therapeutic proteins. More importantly, 11H1 showed enhanced brain penetration after systemic intravenous dosing in a mouse study, which confirmed its potential function as BBB transporter for therapeutic proteins.","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41635901","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Determination of nucleoside DOT1L inhibitors’ residence times by τRAMD simulations τRAMD模拟测定核苷DOT1L抑制剂的停留时间
Pub Date : 2023-01-09 DOI: 10.3389/fddsv.2022.1083198
Carlos D. Flores-León, Luis Fernando Colorado-Pablo, Miguel Á. Santos-Contreras, R. Aguayo‐Ortiz
Human epigenetic enzyme disruptor of telomeric silencing 1-like (DOT1L) is a key drug target for treating acute myeloid leukemia. Several nucleoside and non-nucleoside DOT1L inhibitors have been developed to inhibit its histone methyltransferase activity. Non-mechanism-based nucleoside DOT1L inhibitors have shown good inhibitory activity and high on-target residence times. Previous computational studies have explored the dynamic behavior of this group of molecules on DOT1L to design compounds with enhanced binding affinities. Nevertheless, it is well known that drug-target kinetics also plays a crucial role in the discovery of new drugs. Therefore, we performed τ-Random Acceleration Molecular Dynamics (τRAMD) simulations to estimate the residence times of nucleoside DOT1L inhibitors. The high correlation between the calculated and experimental residence times suggested that the method can reliably estimate the residence time of nucleoside DOT1L inhibitors when modifications are made to those substituents that occupy the buried hydrophobic pocket of the active site, exhibit hydrophobic interactions with F245 or that form H-bonds with D161 and G163. Overall, this study will be a step toward understanding the binding kinetics of nucleoside DOT1L inhibitors for the treatment of acute myeloid leukemia.
人表观遗传学端粒沉默1-样酶干扰因子(DOT1L)是治疗急性粒细胞白血病的关键药物靶点。已经开发了几种核苷和非核苷DOT1L抑制剂来抑制其组蛋白甲基转移酶活性。基于非机制的核苷DOT1L抑制剂显示出良好的抑制活性和高的靶向停留时间。先前的计算研究已经探索了这组分子在DOT1L上的动力学行为,以设计具有增强结合亲和力的化合物。然而,众所周知,药物靶向动力学在新药的发现中也起着至关重要的作用。因此,我们进行了τ-随机加速分子动力学(τRAMD)模拟,以估计核苷DOT1L抑制剂的停留时间。计算的停留时间和实验的停留时间之间的高度相关性表明,当对那些占据活性位点的隐藏疏水口袋、与F245表现出疏水相互作用或与D161和G163形成氢键的取代基进行修饰时,该方法可以可靠地估计核苷DOT1L抑制剂的停留时间。总的来说,这项研究将是了解核苷DOT1L抑制剂治疗急性粒细胞白血病的结合动力学的一步。
{"title":"Determination of nucleoside DOT1L inhibitors’ residence times by τRAMD simulations","authors":"Carlos D. Flores-León, Luis Fernando Colorado-Pablo, Miguel Á. Santos-Contreras, R. Aguayo‐Ortiz","doi":"10.3389/fddsv.2022.1083198","DOIUrl":"https://doi.org/10.3389/fddsv.2022.1083198","url":null,"abstract":"Human epigenetic enzyme disruptor of telomeric silencing 1-like (DOT1L) is a key drug target for treating acute myeloid leukemia. Several nucleoside and non-nucleoside DOT1L inhibitors have been developed to inhibit its histone methyltransferase activity. Non-mechanism-based nucleoside DOT1L inhibitors have shown good inhibitory activity and high on-target residence times. Previous computational studies have explored the dynamic behavior of this group of molecules on DOT1L to design compounds with enhanced binding affinities. Nevertheless, it is well known that drug-target kinetics also plays a crucial role in the discovery of new drugs. Therefore, we performed τ-Random Acceleration Molecular Dynamics (τRAMD) simulations to estimate the residence times of nucleoside DOT1L inhibitors. The high correlation between the calculated and experimental residence times suggested that the method can reliably estimate the residence time of nucleoside DOT1L inhibitors when modifications are made to those substituents that occupy the buried hydrophobic pocket of the active site, exhibit hydrophobic interactions with F245 or that form H-bonds with D161 and G163. Overall, this study will be a step toward understanding the binding kinetics of nucleoside DOT1L inhibitors for the treatment of acute myeloid leukemia.","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48062743","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Drug repurposing screening validated by experimental assays identifies two clinical drugs targeting SARS-CoV-2 main protease 经实验验证的药物再利用筛选鉴定出两种靶向SARS-CoV-2主要蛋白酶的临床药物
Pub Date : 2023-01-05 DOI: 10.3389/fddsv.2022.1082065
Denis N Prada Gori, S. Ruatta, Martín Fló, L. Alberca, C. Bellera, Soonju Park, Jinyeong Heo, Honggun Lee, K. P. Park, O. Pritsch, D. Shum, M. Comini, A. Talevi
The COVID-19 pandemic prompted several drug repositioning initiatives with the aim to rapidly deliver pharmacological candidates able to reduce SARS-CoV-2 dissemination and mortality. A major issue shared by many of the in silico studies addressing the discovery of compounds or drugs targeting SARS-CoV-2 molecules is that they lacked experimental validation of the results. Here we present a computer-aided drug-repositioning campaign against the indispensable SARS-CoV-2 main protease (MPro or 3CLPro) that involved the development of ligand-based ensemble models and the experimental testing of a small subset of the identified hits. The search method explored random subspaces of molecular descriptors to obtain linear classifiers. The best models were then combined by selective ensemble learning to improve their predictive power. Both the individual models and the ensembles were validated by retrospective screening, and later used to screen the DrugBank, Drug Repurposing Hub and Sweetlead libraries for potential inhibitors of MPro. From the 4 in silico hits assayed, atpenin and tinostamustine inhibited MPro (IC50 1 µM and 4 μM, respectively) but not the papain-like protease of SARS-CoV-2 (drugs tested at 25 μM). Preliminary kinetic characterization suggests that tinostamustine and atpenin inhibit MPro by an irreversible and acompetitive mechanisms, respectively. Both drugs failed to inhibit the proliferation of SARS-CoV-2 in VERO cells. The virtual screening method reported here may be a powerful tool to further extent the identification of novel MPro inhibitors. Furthermore, the confirmed MPro hits may be subjected to optimization or retrospective search strategies to improve their molecular target and anti-viral potency.
COVID-19大流行促使了几项药物重新定位举措,目的是快速提供能够减少SARS-CoV-2传播和死亡率的候选药物。许多关于发现针对SARS-CoV-2分子的化合物或药物的计算机研究的一个主要问题是,它们缺乏对结果的实验验证。在这里,我们提出了一种针对必不可少的SARS-CoV-2主要蛋白酶(MPro或3CLPro)的计算机辅助药物重新定位运动,该运动涉及基于配体的集合模型的开发和对已确定的一小部分命中的实验测试。该搜索方法对分子描述符的随机子空间进行搜索,得到线性分类器。然后通过选择性集成学习将最佳模型组合起来以提高其预测能力。通过回顾性筛选验证了个体模型和集合,随后用于筛选DrugBank、Drug Repurposing Hub和Sweetlead库中潜在的MPro抑制剂。在4个实验中,atpenin和tinostamustine抑制MPro (IC50分别为1 μM和4 μM),但对SARS-CoV-2的木瓜蛋白酶没有抑制作用(药物测试在25 μM)。初步的动力学表征表明,丁司莫司汀和atpenin分别通过不可逆机制和竞争机制抑制MPro。这两种药物都未能抑制SARS-CoV-2在VERO细胞中的增殖。本文报道的虚拟筛选方法可能是进一步鉴定新型MPro抑制剂的有力工具。此外,已确认的MPro命中可以进行优化或回顾性搜索策略,以提高其分子靶点和抗病毒效力。
{"title":"Drug repurposing screening validated by experimental assays identifies two clinical drugs targeting SARS-CoV-2 main protease","authors":"Denis N Prada Gori, S. Ruatta, Martín Fló, L. Alberca, C. Bellera, Soonju Park, Jinyeong Heo, Honggun Lee, K. P. Park, O. Pritsch, D. Shum, M. Comini, A. Talevi","doi":"10.3389/fddsv.2022.1082065","DOIUrl":"https://doi.org/10.3389/fddsv.2022.1082065","url":null,"abstract":"The COVID-19 pandemic prompted several drug repositioning initiatives with the aim to rapidly deliver pharmacological candidates able to reduce SARS-CoV-2 dissemination and mortality. A major issue shared by many of the in silico studies addressing the discovery of compounds or drugs targeting SARS-CoV-2 molecules is that they lacked experimental validation of the results. Here we present a computer-aided drug-repositioning campaign against the indispensable SARS-CoV-2 main protease (MPro or 3CLPro) that involved the development of ligand-based ensemble models and the experimental testing of a small subset of the identified hits. The search method explored random subspaces of molecular descriptors to obtain linear classifiers. The best models were then combined by selective ensemble learning to improve their predictive power. Both the individual models and the ensembles were validated by retrospective screening, and later used to screen the DrugBank, Drug Repurposing Hub and Sweetlead libraries for potential inhibitors of MPro. From the 4 in silico hits assayed, atpenin and tinostamustine inhibited MPro (IC50 1 µM and 4 μM, respectively) but not the papain-like protease of SARS-CoV-2 (drugs tested at 25 μM). Preliminary kinetic characterization suggests that tinostamustine and atpenin inhibit MPro by an irreversible and acompetitive mechanisms, respectively. Both drugs failed to inhibit the proliferation of SARS-CoV-2 in VERO cells. The virtual screening method reported here may be a powerful tool to further extent the identification of novel MPro inhibitors. Furthermore, the confirmed MPro hits may be subjected to optimization or retrospective search strategies to improve their molecular target and anti-viral potency.","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41404972","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Editorial: Insights in silico methods and artificial intelligence for drug discovery: 2022 社论:对药物发现的硅方法和人工智能的见解:2022
Pub Date : 2023-01-05 DOI: 10.3389/fddsv.2022.1126955
J. Medina‐Franco
Entering the third decade of the 21st Century, artificial intelligence (AI) continues to offer significant advances in drug discovery (Jiménez-Luna et al., 2021; Jayatunga et al., 2022). When used rationally beyond the hype, AI offers clear promise to advance further basic and applied research (Medina-Franco et al., 2021). At the same time, AI faces challenges to address at different levels. The present Research Topic brings together experts worldwide from industry, academic, not-for-profit, and governmental settings to openly discuss novel insights, recent advances, latest discoveries, and current challenges in the field of In silico Methods and Artificial Intelligence for Drug Discovery. From an industry point of view, DiNuzzo presents a perspective on how AI enables the modeling and simulation of biological networks to accelerate drug discovery. He emphasizes that the proper combination of the predictive capability of AI with the mechanistic knowledge of modeling and simulation is expected to provide a major contribution to the pharmaceutical industry. DiNuzzo also concludes that AI will be a key player in analyzing biological networks that will deliver substantial progress towards the improvement of drug target identification and validation, qualify potentially associated side-effects, identify the efficacy and toxicity of biomarkers, aid with hypothesis generation, optimal experimental design, and testing for disease understanding and identification of disease biomarkers. McDermott et al. discuss a platform based on AI that aids in the discovery of DNA damaging agents for ultra-rare cancer atypical teratoid rhabdoid tumors (ATRT). Specifically, the authors showed the power of using the public USA’s National Cancer Institute (NCI)’s CellMiner Cross Database and Lantern Pharma’s proprietary AI and machine learning (ML) RADR® platform to uncover biological insights and potentially new target indications for the acylfulvene derivative drugs LP-100 (Irofulven) and LP-184. Lantern’s AI and ML RADR® platform was used to develop a model to test, computationally, if LP-184 would be effective in ATRT patients. RADR® suggested that ATRT would be sensitive to LP-184, which was then validated in vitro and in vivo. Namba-Nzanguim et al. review how AI is helping to advance antiviral drug discovery in low-resourced settings. Authors shared their perspectives on the benefits, limitations, and pitfalls of AI/ML tools in the discovery of novel antivirals. Namba-Nzanguim et al. also present current and novel data sharing models, including intellectual property-preserving AI/ML. Authors concluded that AI/ML provides a cost-effective solution for developing antivirals, but AI/ML tools depend on improved access to viral assays data and better data integration protocols. Schmitz et al. OPEN ACCESS
进入21世纪的第三个十年,人工智能(AI)继续在药物发现方面取得重大进展(Jiménez-Luna等人,2021;Jayatunga等人,2022)。当在炒作之外合理使用时,人工智能为进一步推进基础和应用研究提供了明确的承诺(Medina Franco et al.,2021)。与此同时,人工智能面临着不同层面的挑战。本研究主题汇集了来自世界各地行业、学术界、非营利组织和政府机构的专家,公开讨论药物发现的计算机方法和人工智能领域的新见解、最新进展、最新发现和当前挑战。从行业的角度来看,DiNuzzo介绍了人工智能如何使生物网络的建模和模拟加速药物发现。他强调,人工智能的预测能力与建模和模拟的机械知识的适当结合有望为制药行业做出重大贡献。DiNuzzo还得出结论,人工智能将在分析生物网络方面发挥关键作用,该网络将在改进药物靶点识别和验证、鉴定潜在的相关副作用、识别生物标志物的疗效和毒性、帮助产生假设、优化实验设计、,以及测试疾病理解和疾病生物标志物的鉴定。McDermott等人讨论了一个基于人工智能的平台,该平台有助于发现超恶性癌症非典型畸胎瘤样横纹肌样肿瘤(ATRT)的DNA损伤剂。具体而言,作者展示了使用美国国家癌症研究所(NCI)的CellMiner交叉数据库和Lantern Pharma专有的人工智能和机器学习(ML)RADR®平台来揭示酰基富烯衍生物药物LP-100(Irofulven)和LP-184的生物学见解和潜在新靶点适应症的力量。Lantern的AI和ML RADR®平台用于开发一个模型,通过计算测试LP-184是否对ATRT患者有效。RADR®表明ATRT对LP-184敏感,随后在体外和体内进行了验证。Namba Nzanguim等人综述了人工智能如何在资源匮乏的环境中帮助推进抗病毒药物的发现。作者分享了他们对AI/ML工具在发现新型抗病毒药物方面的好处、局限性和陷阱的看法。Namba Nzanguim等人还介绍了当前和新的数据共享模型,包括保护知识产权的AI/ML。作者得出结论,AI/ML为开发抗病毒药物提供了一种具有成本效益的解决方案,但AI/ML工具依赖于改进对病毒检测数据的访问和更好的数据集成协议。Schmitz等人开放访问
{"title":"Editorial: Insights in silico methods and artificial intelligence for drug discovery: 2022","authors":"J. Medina‐Franco","doi":"10.3389/fddsv.2022.1126955","DOIUrl":"https://doi.org/10.3389/fddsv.2022.1126955","url":null,"abstract":"Entering the third decade of the 21st Century, artificial intelligence (AI) continues to offer significant advances in drug discovery (Jiménez-Luna et al., 2021; Jayatunga et al., 2022). When used rationally beyond the hype, AI offers clear promise to advance further basic and applied research (Medina-Franco et al., 2021). At the same time, AI faces challenges to address at different levels. The present Research Topic brings together experts worldwide from industry, academic, not-for-profit, and governmental settings to openly discuss novel insights, recent advances, latest discoveries, and current challenges in the field of In silico Methods and Artificial Intelligence for Drug Discovery. From an industry point of view, DiNuzzo presents a perspective on how AI enables the modeling and simulation of biological networks to accelerate drug discovery. He emphasizes that the proper combination of the predictive capability of AI with the mechanistic knowledge of modeling and simulation is expected to provide a major contribution to the pharmaceutical industry. DiNuzzo also concludes that AI will be a key player in analyzing biological networks that will deliver substantial progress towards the improvement of drug target identification and validation, qualify potentially associated side-effects, identify the efficacy and toxicity of biomarkers, aid with hypothesis generation, optimal experimental design, and testing for disease understanding and identification of disease biomarkers. McDermott et al. discuss a platform based on AI that aids in the discovery of DNA damaging agents for ultra-rare cancer atypical teratoid rhabdoid tumors (ATRT). Specifically, the authors showed the power of using the public USA’s National Cancer Institute (NCI)’s CellMiner Cross Database and Lantern Pharma’s proprietary AI and machine learning (ML) RADR® platform to uncover biological insights and potentially new target indications for the acylfulvene derivative drugs LP-100 (Irofulven) and LP-184. Lantern’s AI and ML RADR® platform was used to develop a model to test, computationally, if LP-184 would be effective in ATRT patients. RADR® suggested that ATRT would be sensitive to LP-184, which was then validated in vitro and in vivo. Namba-Nzanguim et al. review how AI is helping to advance antiviral drug discovery in low-resourced settings. Authors shared their perspectives on the benefits, limitations, and pitfalls of AI/ML tools in the discovery of novel antivirals. Namba-Nzanguim et al. also present current and novel data sharing models, including intellectual property-preserving AI/ML. Authors concluded that AI/ML provides a cost-effective solution for developing antivirals, but AI/ML tools depend on improved access to viral assays data and better data integration protocols. Schmitz et al. OPEN ACCESS","PeriodicalId":73080,"journal":{"name":"Frontiers in drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43510989","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Frontiers in drug discovery
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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