Pub Date : 2025-12-01Epub Date: 2025-11-25DOI: 10.1080/17460441.2025.2593382
Rongchao Wang, Lihua Yang, Lei Du, Li Zhao, Siyu Chen, Weihu Li, Daoxin Dai, Binhai Shi, Jingli Xie
Background: Skin aging is linked to the overactivity of matrix metalloproteinases (MMPs) and elastase, making their inhibition a promising approach for antiaging. This study aimed to discover novel antiaging peptides from Chlorella proteins using high-throughput virtual screening.
Methods: Batch molecular docking protocol with a custom Python script for 3D peptide structure modeling and AutoDock Vina was applied to predict inhibitory peptides on MMPs and elastase from 1,965 peptides theoretically resistant to gastrointestinal digestion. The top candidates were synthesized for activity assay, and MD simulation illustrated the binding mechanism of potent peptides.
Results: Seventeen peptides with a binding energy < -7.0 kcal/mol showed IC50 ≤ 150 μM. Peptide DGSY acted high potency against MMP-1 (IC50 = 32.6 μM), and HDISHW inhibited MMP-9 and elastase at the lowest IC50 (20.1, 16.5 μM). GAASF inhibited all three enzymes (IC50 = 54.0, 41.9, 62.5 μM). MD simulations confirmed the stability of these peptide-protein complexes, which coincided with the in vitro activity well.
Conclusion: The virtual strategy efficiently identified multifunctional antiaging peptides and could accelerate the discovery of bioactive peptides for cosmetic and therapeutic use. Additionally, its efficiency makes it useful for building high-quality training sets in deep learning models for bioactive structure discovery.
{"title":"Discovery of novel inhibitory peptides on matrix metalloproteinases and elastase for skin antiaging using batch molecular docking strategy.","authors":"Rongchao Wang, Lihua Yang, Lei Du, Li Zhao, Siyu Chen, Weihu Li, Daoxin Dai, Binhai Shi, Jingli Xie","doi":"10.1080/17460441.2025.2593382","DOIUrl":"10.1080/17460441.2025.2593382","url":null,"abstract":"<p><strong>Background: </strong>Skin aging is linked to the overactivity of matrix metalloproteinases (MMPs) and elastase, making their inhibition a promising approach for antiaging. This study aimed to discover novel antiaging peptides from Chlorella proteins using high-throughput virtual screening.</p><p><strong>Methods: </strong>Batch molecular docking protocol with a custom Python script for 3D peptide structure modeling and AutoDock Vina was applied to predict inhibitory peptides on MMPs and elastase from 1,965 peptides theoretically resistant to gastrointestinal digestion. The top candidates were synthesized for activity assay, and MD simulation illustrated the binding mechanism of potent peptides.</p><p><strong>Results: </strong>Seventeen peptides with a binding energy < -7.0 kcal/mol showed IC<sub>50</sub> ≤ 150 μM. Peptide DGSY acted high potency against MMP-1 (IC<sub>50</sub> = 32.6 μM), and HDISHW inhibited MMP-9 and elastase at the lowest IC<sub>50</sub> (20.1, 16.5 μM). GAASF inhibited all three enzymes (IC<sub>50</sub> = 54.0, 41.9, 62.5 μM). MD simulations confirmed the stability of these peptide-protein complexes, which coincided with the in vitro activity well.</p><p><strong>Conclusion: </strong>The virtual strategy efficiently identified multifunctional antiaging peptides and could accelerate the discovery of bioactive peptides for cosmetic and therapeutic use. Additionally, its efficiency makes it useful for building high-quality training sets in deep learning models for bioactive structure discovery.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1713-1724"},"PeriodicalIF":4.9,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145563299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-24DOI: 10.1080/17460441.2025.2578001
Aditi Gangopadhyay, Abhijit Datta
Introduction: The recent surge in cholera outbreaks worldwide, partly driven by climate change, highlights its potential as a significant public health threat. The absence of definitive treatments underscores the urgent need for developing effective targeted therapeutics. The cholera holotoxin comprises a catalytically active A1 subunit, which mediates ADP-ribosylation to induce secretory diarrhea, and a pentameric B subunit responsible for toxin-host cell attachment via GM1 receptors. A1 activation requires binding to human ADP-ribosylation factor 6 (ARF6). Although the inhibition of B-pentamer - GM1 binding has been extensively investigated, several structural and pharmacokinetic challenges remain.
Areas covered: This article is based on a keyword-based literature survey across relevant research repositories, covering studies published up to 2025. It summarizes the structure- and ligand-based molecular modeling approaches employed for identifying inhibitors targeting toxin-host binding, including GM1 mimetics, glycomimetics, and natural compounds. Alternative avenues of toxin inhibition, including occlusion of the B-pentamer pore, A1 catalytic site, and the A1-ARF6 interface to disrupt toxin assembly, ADP-ribosylation, and A1 activation, respectively, are also discussed.
Expert opinion: Targeting the B-pentamer pore, A1 active site, or A1-ARF6 interface holds significant therapeutic potential against cholera-induced dehydration and hypovolaemic shock. These underexplored yet promising druggable targets warrant further investigation for developing effective, targeted therapies.
{"title":"Advances in cholera toxin inhibitor design: insights from molecular modelling.","authors":"Aditi Gangopadhyay, Abhijit Datta","doi":"10.1080/17460441.2025.2578001","DOIUrl":"10.1080/17460441.2025.2578001","url":null,"abstract":"<p><strong>Introduction: </strong>The recent surge in cholera outbreaks worldwide, partly driven by climate change, highlights its potential as a significant public health threat. The absence of definitive treatments underscores the urgent need for developing effective targeted therapeutics. The cholera holotoxin comprises a catalytically active A1 subunit, which mediates ADP-ribosylation to induce secretory diarrhea, and a pentameric B subunit responsible for toxin-host cell attachment via GM1 receptors. A1 activation requires binding to human ADP-ribosylation factor 6 (ARF6). Although the inhibition of B-pentamer - GM1 binding has been extensively investigated, several structural and pharmacokinetic challenges remain.</p><p><strong>Areas covered: </strong>This article is based on a keyword-based literature survey across relevant research repositories, covering studies published up to 2025. It summarizes the structure- and ligand-based molecular modeling approaches employed for identifying inhibitors targeting toxin-host binding, including GM1 mimetics, glycomimetics, and natural compounds. Alternative avenues of toxin inhibition, including occlusion of the B-pentamer pore, A1 catalytic site, and the A1-ARF6 interface to disrupt toxin assembly, ADP-ribosylation, and A1 activation, respectively, are also discussed.</p><p><strong>Expert opinion: </strong>Targeting the B-pentamer pore, A1 active site, or A1-ARF6 interface holds significant therapeutic potential against cholera-induced dehydration and hypovolaemic shock. These underexplored yet promising druggable targets warrant further investigation for developing effective, targeted therapies.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1607-1619"},"PeriodicalIF":4.9,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145354317","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-30DOI: 10.1080/17460441.2025.2594642
Piet Geusens, Wim Van Hul, Joop van den Bergh, Willem Lems
Introduction: The discovery of romosozumab, a monoclonal antibody to sclerostin and treatment option for severe osteoporosis, resulted from convergent genetic research of persons with rare hyperostotic bone diseases and the discovery of the Wnt-signaling pathway, a vital pathway in bone metabolism.
Areas covered: The authors provide an overview of the discovery of the SOST gene in humans and of Wnt signaling in animals, leading to the identification of sclerostin, a major regulator of bone formation and resorption. The authors further provide an overview of the studies that led to the development of romosozumab, a unique dual action monoclonal antibody that increases bone formation while decreasing bone resorption.
Expert opinion: In postmenopausal women, the administration of romosozumab over one year decreased the risk of vertebral and clinical fractures versus placebo and versus alendronate. Furthermore, sequential treatment, switching romosozumab over to denosumab, reduced the risk of vertebral fractures compared to switching the placebo to denosumab. Meanwhile, switching romosozumab to alendronate reduced the risk of vertebral, clinical, nonvertebral, and hip fractures compared to continuous alendronate. An imbalance in cardiovascular events was found when using romosozumab in comparison to alendronate but not versus placebo. Romosozumab was eventually approved by EMA and FDA in 2019 for the treatment of patients with very high risk of fractures while considering their cardiovascular risk and is available and reimbursed in many countries.
{"title":"The preclinical discovery and development of romosozumab for the treatment of people with severe osteoporosis who are at high risk of fracture.","authors":"Piet Geusens, Wim Van Hul, Joop van den Bergh, Willem Lems","doi":"10.1080/17460441.2025.2594642","DOIUrl":"10.1080/17460441.2025.2594642","url":null,"abstract":"<p><strong>Introduction: </strong>The discovery of romosozumab, a monoclonal antibody to sclerostin and treatment option for severe osteoporosis, resulted from convergent genetic research of persons with rare hyperostotic bone diseases and the discovery of the Wnt-signaling pathway, a vital pathway in bone metabolism.</p><p><strong>Areas covered: </strong>The authors provide an overview of the discovery of the <i>SOST</i> gene in humans and of Wnt signaling in animals, leading to the identification of sclerostin, a major regulator of bone formation and resorption. The authors further provide an overview of the studies that led to the development of romosozumab, a unique dual action monoclonal antibody that increases bone formation while decreasing bone resorption.</p><p><strong>Expert opinion: </strong>In postmenopausal women, the administration of romosozumab over one year decreased the risk of vertebral and clinical fractures versus placebo and versus alendronate. Furthermore, sequential treatment, switching romosozumab over to denosumab, reduced the risk of vertebral fractures compared to switching the placebo to denosumab. Meanwhile, switching romosozumab to alendronate reduced the risk of vertebral, clinical, nonvertebral, and hip fractures compared to continuous alendronate. An imbalance in cardiovascular events was found when using romosozumab in comparison to alendronate but not versus placebo. Romosozumab was eventually approved by EMA and FDA in 2019 for the treatment of patients with very high risk of fractures while considering their cardiovascular risk and is available and reimbursed in many countries.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1475-1492"},"PeriodicalIF":4.9,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145596370","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1080/17460441.2025.2594641
Hikaru Ishikura, James A Bull
Introduction: Oxetanes are increasingly prevalent motifs in medicinal chemistry. While oxetanes feature famously in the taxol family, it was not until the recent approval of rilzabrutinib that they have been validated in a fully synthetic drug. These four-membered oxygen-containing rings are valued for their ability to modulate key physicochemical properties such as polarity, lipophilicity, and metabolic stability. Their incorporation can improve aqueous solubility, reduce metabolic liability, reduce basicity of adjacent amines, and redirect metabolic clearance away from cytochrome P450 enzymes.
Areas covered: This review examines the role of the oxetane in nine clinical candidates, one very recently approved, two recently discontinued clinical candidates, two tool compounds, and three lead compounds disclosed in 2024. Recent advances indexed in SciFinder-n (2017-2024) in the process-scale and laboratory-scale synthetic routes are discussed, highlighting ongoing innovation required to access these motifs efficiently and sustainably.
Expert opinion: The regulatory approval of rilzabrutinib and likely approval of ziresovir provide confidence in using oxetanes as important elements in drug design. While oxetanes have so far been incorporated primarily as pendant groups to optimize physicochemical properties, their use as scaffolding and binding elements presents an exciting opportunity. Enhanced synthetic accessibility to oxetane derivatives will expedite their inclusion in drug discovery campaigns.
{"title":"Synthetic oxetanes in drug discovery: where are we in 2025?","authors":"Hikaru Ishikura, James A Bull","doi":"10.1080/17460441.2025.2594641","DOIUrl":"10.1080/17460441.2025.2594641","url":null,"abstract":"<p><strong>Introduction: </strong>Oxetanes are increasingly prevalent motifs in medicinal chemistry. While oxetanes feature famously in the taxol family, it was not until the recent approval of rilzabrutinib that they have been validated in a fully synthetic drug. These four-membered oxygen-containing rings are valued for their ability to modulate key physicochemical properties such as polarity, lipophilicity, and metabolic stability. Their incorporation can improve aqueous solubility, reduce metabolic liability, reduce basicity of adjacent amines, and redirect metabolic clearance away from cytochrome P450 enzymes.</p><p><strong>Areas covered: </strong>This review examines the role of the oxetane in nine clinical candidates, one very recently approved, two recently discontinued clinical candidates, two tool compounds, and three lead compounds disclosed in 2024. Recent advances indexed in SciFinder-n (2017-2024) in the process-scale and laboratory-scale synthetic routes are discussed, highlighting ongoing innovation required to access these motifs efficiently and sustainably.</p><p><strong>Expert opinion: </strong>The regulatory approval of rilzabrutinib and likely approval of ziresovir provide confidence in using oxetanes as important elements in drug design. While oxetanes have so far been incorporated primarily as pendant groups to optimize physicochemical properties, their use as scaffolding and binding elements presents an exciting opportunity. Enhanced synthetic accessibility to oxetane derivatives will expedite their inclusion in drug discovery campaigns.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1621-1638"},"PeriodicalIF":4.9,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145654152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-19DOI: 10.1080/17460441.2025.2582539
Sandeep Sundriyal, Animesh Majumdar
Introduction: Malaria remains a major health challenge, with increasing resistance to frontline chemotherapy. Recent cheminformatics studies have revealed that potent antiplasmodials occupy a distinct antimalarial chemical space (AMCS), defined by specific property cutoffs.
Areas covered: We show that only ~ 6-8% of compounds in the representative commercial libraries are AMCS-compliant, emphasizing the need for pre-filtering to improve their suitability for phenotypic screening. Most clinically used antimalarials and several clinically used oral drugs repurposed for antimalarial activity comply with AMCS indicating the important role AMCS can play in rationalizing drug repurposing studies. Furthermore, AMCS-guided scaffold modification offers a valuable strategy during lead optimization. With the help of recent literature examples we show how AMCS can be exploited for multimodal antimalarials or explain confounding structure-activity relationships.
Expert opinion: Due to the lack of funding and economically weaker patient population new antimalarials need to be cost-effective. The concept of AMCS can be harnessed to achieve this goal. Although there are inherent limitations associated with using hard cut offs and existing computational methodologies, the AMCS has strong potential to identify novel chemical matter for designing antimalarials that are resilient to resistance development.
{"title":"Harnessing antimalarial chemical space: the way forward.","authors":"Sandeep Sundriyal, Animesh Majumdar","doi":"10.1080/17460441.2025.2582539","DOIUrl":"10.1080/17460441.2025.2582539","url":null,"abstract":"<p><strong>Introduction: </strong>Malaria remains a major health challenge, with increasing resistance to frontline chemotherapy. Recent cheminformatics studies have revealed that potent antiplasmodials occupy a distinct antimalarial chemical space (AMCS), defined by specific property cutoffs.</p><p><strong>Areas covered: </strong>We show that only ~ 6-8% of compounds in the representative commercial libraries are AMCS-compliant, emphasizing the need for pre-filtering to improve their suitability for phenotypic screening. Most clinically used antimalarials and several clinically used oral drugs repurposed for antimalarial activity comply with AMCS indicating the important role AMCS can play in rationalizing drug repurposing studies. Furthermore, AMCS-guided scaffold modification offers a valuable strategy during lead optimization. With the help of recent literature examples we show how AMCS can be exploited for multimodal antimalarials or explain confounding structure-activity relationships.</p><p><strong>Expert opinion: </strong>Due to the lack of funding and economically weaker patient population new antimalarials need to be cost-effective. The concept of AMCS can be harnessed to achieve this goal. Although there are inherent limitations associated with using hard cut offs and existing computational methodologies, the AMCS has strong potential to identify novel chemical matter for designing antimalarials that are resilient to resistance development.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1513-1524"},"PeriodicalIF":4.9,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145376823","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-27DOI: 10.1080/17460441.2025.2579122
Benjamin R Hoy, Douer Zhu, Nicholas A Veldhuis, Rainer V Haberberger, Nicolas H Voelcker, Dusan Matusica
{"title":"The potential of microfluidic platforms for neuron differentiation and pain modeling in novel drug discovery.","authors":"Benjamin R Hoy, Douer Zhu, Nicholas A Veldhuis, Rainer V Haberberger, Nicolas H Voelcker, Dusan Matusica","doi":"10.1080/17460441.2025.2579122","DOIUrl":"10.1080/17460441.2025.2579122","url":null,"abstract":"","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1469-1473"},"PeriodicalIF":4.9,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145354311","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-12-07DOI: 10.1080/17460441.2025.2599178
Federica Cernuto, Avisa Maleki, Giulia Russo, Valentina Di Salvatore, Francesco Pappalardo
Introduction: In silico technologies are increasingly shaping vaccine development, supporting the field beyond empirical discovery toward rational, data-driven design. Contemporary computational pipelines enable rapid antigen screening, high-precision epitope-MHC binding prediction, structural modeling, and immune response simulations. These approaches are accelerating vaccine discovery not only for infectious diseases but also in oncology, where neoantigen prediction underpins personalized cancer immunotherapy.
Areas covered: This review explores recent advances in computational pipelines for epitope-based vaccine design, covering antigen discovery; B- and T-cell epitope mapping; safety and specificity assessment; vaccine construct assembly with adjuvants and linkers; structural modeling; and immune-response simulations that predict efficacy in specific disease contexts using advanced platforms. It showcases applications in infectious diseases, including SARS-CoV-2, tuberculosis, and influenza, and poxivirus infections, as well as in cancer immunotherapy. It is based on literature obtained through searches utilizing PubMed, Scopus, and Web of Science databases covering publications up to 2025, using combinations of keywords such as epitope-based vaccines, reverse vaccinology, immunoinformatics, and immune system simulation.
Expert opinion: In silico approaches offer a transformative advantage to vaccine research by delivering speed, cost-efficiency, and enhanced precision. Yet the predictive power of current computational pipelines is still constrained by algorithmic limitations and by their incomplete integration of immune-regulatory processes. Progress in artificial intelligence, multi-omics integration, and formal recognition of digital evidence by regulatory agencies will be crucial for narrowing the gap between computational predictions and experimental validation. Ultimately, combining predictive immunoinformatics with advanced immune simulations and rigorous verification could help establish in silico methodologies as a cornerstone of next-generation vaccine development.
引言:在硅技术越来越多地塑造疫苗的发展,支持领域超越经验发现走向理性,数据驱动的设计。现代计算管道能够实现快速抗原筛选,高精度表位- mhc结合预测,结构建模和免疫反应模拟。这些方法不仅加速了传染病疫苗的发现,而且加速了肿瘤学疫苗的发现,在肿瘤学领域,新抗原预测是个性化癌症免疫治疗的基础。涵盖领域:本综述探讨了基于表位的疫苗设计的计算管道的最新进展,包括抗原发现;B细胞和t细胞表位定位;安全性和特异性评价;含佐剂和连接剂的疫苗构建组装;结构建模;以及利用先进平台预测特定疾病情况下疗效的免疫反应模拟。它展示了在传染性疾病(包括SARS-CoV-2、结核病和流感)和痘病毒感染以及癌症免疫治疗中的应用。它基于通过PubMed、Scopus和Web of Science数据库检索获得的文献,涵盖截至2025年的出版物,使用诸如基于表位的疫苗、反向疫苗学、免疫信息学和免疫系统模拟等关键词的组合。专家意见:计算机方法通过提供速度、成本效益和更高的精度,为疫苗研究提供了变革性优势。然而,当前计算管道的预测能力仍然受到算法限制以及它们对免疫调节过程的不完整整合的限制。人工智能、多组学整合和监管机构对数字证据的正式认可方面的进展,对于缩小计算预测和实验验证之间的差距至关重要。最终,将预测免疫信息学与先进的免疫模拟和严格的验证相结合,可以帮助建立计算机方法,作为下一代疫苗开发的基石。
{"title":"<i>In-silico</i> epitope-based vaccines design: progress, challenges and the road ahead.","authors":"Federica Cernuto, Avisa Maleki, Giulia Russo, Valentina Di Salvatore, Francesco Pappalardo","doi":"10.1080/17460441.2025.2599178","DOIUrl":"10.1080/17460441.2025.2599178","url":null,"abstract":"<p><strong>Introduction: </strong>In silico technologies are increasingly shaping vaccine development, supporting the field beyond empirical discovery toward rational, data-driven design. Contemporary computational pipelines enable rapid antigen screening, high-precision epitope-MHC binding prediction, structural modeling, and immune response simulations. These approaches are accelerating vaccine discovery not only for infectious diseases but also in oncology, where neoantigen prediction underpins personalized cancer immunotherapy.</p><p><strong>Areas covered: </strong>This review explores recent advances in computational pipelines for epitope-based vaccine design, covering antigen discovery; B- and T-cell epitope mapping; safety and specificity assessment; vaccine construct assembly with adjuvants and linkers; structural modeling; and immune-response simulations that predict efficacy in specific disease contexts using advanced platforms. It showcases applications in infectious diseases, including SARS-CoV-2, tuberculosis, and influenza, and poxivirus infections, as well as in cancer immunotherapy. It is based on literature obtained through searches utilizing PubMed, Scopus, and Web of Science databases covering publications up to 2025, using combinations of keywords such as epitope-based vaccines, reverse vaccinology, immunoinformatics, and immune system simulation.</p><p><strong>Expert opinion: </strong>In silico approaches offer a transformative advantage to vaccine research by delivering speed, cost-efficiency, and enhanced precision. Yet the predictive power of current computational pipelines is still constrained by algorithmic limitations and by their incomplete integration of immune-regulatory processes. Progress in artificial intelligence, multi-omics integration, and formal recognition of digital evidence by regulatory agencies will be crucial for narrowing the gap between computational predictions and experimental validation. Ultimately, combining predictive immunoinformatics with advanced immune simulations and rigorous verification could help establish in silico methodologies as a cornerstone of next-generation vaccine development.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1701-1712"},"PeriodicalIF":4.9,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145700313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-09-23DOI: 10.1080/17460441.2025.2562017
Albert Adell
Introduction: N-methyl-D-aspartate (NMDA) receptor antagonists such as phencyclidine (PCP) and ketamine can induce schizophrenic features in healthy volunteers and exacerbate the symptoms in schizophrenic patients. Furthermore, the administration of NMDA receptor antagonists to rodents produces hyperlocomotion. The ability of drugs to attenuate this hyperlocomotion correlates with clinical efficacy on positive symptoms. Similarly, social withdrawal is taken as a surrogate of unsociability in schizophrenia. Furthermore, first episode psychosis and chronic schizophrenia can be modeled by acute and subchronic administration of NMDA receptor blockers, respectively. Therefore, the NMDA hypofunction model provides a powerful tool to develop new therapeutic strategies in drug discovery to treat schizophrenia.
Areas covered: This perspective describes the similitudes between schizophrenia in humans and the traits demonstrated by rodent models based upon the hypofunction of NMDA receptors. Comparisons are made in terms of behavioral, neurochemical, neuroimaging and neurophysiological studies. Different therapeutic responses are also discussed.
Expert opinion: Both schizophrenic patients and developed rodent models exhibit many similitudes such as decreased expression of NMDA receptors, enhanced dopaminergic and serotonergic transmission as well as altered gamma oscillations and deficits in cognitive paradigms. The NMDA receptor antagonism model can thus represent an excellent strategy to study the neurobiological underpinnings of schizophrenia and the potential therapeutic role of new antipsychotic drugs.
n -甲基- d -天冬氨酸(NMDA)受体拮抗剂如苯环利定(PCP)和氯胺酮可在健康志愿者中诱发精神分裂症特征,并加重精神分裂症患者的症状。此外,NMDA受体拮抗剂对啮齿动物产生过度运动。药物减轻这种过度运动的能力与阳性症状的临床疗效相关。同样,社会退缩被认为是精神分裂症中不合群的替代。此外,首发精神病和慢性精神分裂症可以分别通过急性和亚慢性给药NMDA受体阻滞剂来模拟。因此,NMDA功能减退模型为开发治疗精神分裂症的药物开发新策略提供了强有力的工具。涵盖领域:这一视角描述了人类精神分裂症与基于NMDA受体功能低下的啮齿动物模型所展示的特征之间的相似性。在行为学、神经化学、神经影像学和神经生理学研究方面进行了比较。还讨论了不同的治疗反应。专家意见:精神分裂症患者和发达的啮齿动物模型都表现出许多相似之处,如NMDA受体表达减少,多巴胺能和血清素能传递增强,伽马振荡改变和认知范式缺陷。因此,NMDA受体拮抗模型可以为研究精神分裂症的神经生物学基础和新型抗精神病药物的潜在治疗作用提供一个极好的策略。
{"title":"What value do NMDA receptor antagonist models of schizophrenia have for novel drug discovery?","authors":"Albert Adell","doi":"10.1080/17460441.2025.2562017","DOIUrl":"10.1080/17460441.2025.2562017","url":null,"abstract":"<p><strong>Introduction: </strong>N-methyl-D-aspartate (NMDA) receptor antagonists such as phencyclidine (PCP) and ketamine can induce schizophrenic features in healthy volunteers and exacerbate the symptoms in schizophrenic patients. Furthermore, the administration of NMDA receptor antagonists to rodents produces hyperlocomotion. The ability of drugs to attenuate this hyperlocomotion correlates with clinical efficacy on positive symptoms. Similarly, social withdrawal is taken as a surrogate of unsociability in schizophrenia. Furthermore, first episode psychosis and chronic schizophrenia can be modeled by acute and subchronic administration of NMDA receptor blockers, respectively. Therefore, the NMDA hypofunction model provides a powerful tool to develop new therapeutic strategies in drug discovery to treat schizophrenia.</p><p><strong>Areas covered: </strong>This perspective describes the similitudes between schizophrenia in humans and the traits demonstrated by rodent models based upon the hypofunction of NMDA receptors. Comparisons are made in terms of behavioral, neurochemical, neuroimaging and neurophysiological studies. Different therapeutic responses are also discussed.</p><p><strong>Expert opinion: </strong>Both schizophrenic patients and developed rodent models exhibit many similitudes such as decreased expression of NMDA receptors, enhanced dopaminergic and serotonergic transmission as well as altered gamma oscillations and deficits in cognitive paradigms. The NMDA receptor antagonism model can thus represent an excellent strategy to study the neurobiological underpinnings of schizophrenia and the potential therapeutic role of new antipsychotic drugs.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1377-1385"},"PeriodicalIF":4.9,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145069397","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-09-01DOI: 10.1080/17460441.2025.2555275
Alan Talevi, Lucas N Alberca, Carolina L Bellera
Introduction: The search for molecular novelty frequently collides with the fact that drug candidates with the best translational prospects are confined to - or concentrated in - defined regions of chemical space. The new possibilities of AI, particularly retrosynthesis prediction and generative AI, allow for the automated or semi-automated exploration of less restricted and unexplored areas of chemical space.
Areas covered: The notion of novelty in drug discovery is discussed, and representative examples of AI-guided de novo drug design, optimization, and retrosynthesis prediction are presented, with a focus on reports on open-source tools published in the last 3 years (2022-2025). Scopus was used to search relevant literature.
Expert opinion: Modern deep learning architectures have been adapted for the de novo design and molecular optimization. These technologies, and especially those based on conditional generation, will possibly have a great impact on expanding the regions of chemical space that are exploited therapeutically. However, there are some persistent challenges in the field that are gradually being addressed, including how to assess the synthetic accessibility of designed molecules without compromising the generation of structural novelty; the need to increase the availability and diversity of benchmark datasets; and the relative scarcity of large-scale experimental validation of the designs.
{"title":"Tackling the issue of confined chemical space with AI-based de novo drug design and molecular optimization.","authors":"Alan Talevi, Lucas N Alberca, Carolina L Bellera","doi":"10.1080/17460441.2025.2555275","DOIUrl":"10.1080/17460441.2025.2555275","url":null,"abstract":"<p><strong>Introduction: </strong>The search for molecular novelty frequently collides with the fact that drug candidates with the best translational prospects are confined to - or concentrated in - defined regions of chemical space. The new possibilities of AI, particularly retrosynthesis prediction and generative AI, allow for the automated or semi-automated exploration of less restricted and unexplored areas of chemical space.</p><p><strong>Areas covered: </strong>The notion of novelty in drug discovery is discussed, and representative examples of AI-guided de novo drug design, optimization, and retrosynthesis prediction are presented, with a focus on reports on open-source tools published in the last 3 years (2022-2025). Scopus was used to search relevant literature.</p><p><strong>Expert opinion: </strong>Modern deep learning architectures have been adapted for the de novo design and molecular optimization. These technologies, and especially those based on conditional generation, will possibly have a great impact on expanding the regions of chemical space that are exploited therapeutically. However, there are some persistent challenges in the field that are gradually being addressed, including how to assess the synthetic accessibility of designed molecules without compromising the generation of structural novelty; the need to increase the availability and diversity of benchmark datasets; and the relative scarcity of large-scale experimental validation of the designs.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1405-1418"},"PeriodicalIF":4.9,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144948366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-09-15DOI: 10.1080/17460441.2025.2558161
Alberto Marbán-González, Verónica Ramírez-Cid, Alejandro Cristóbal-Ramírez, José L Medina-Franco
Introduction: Cheminformatics has become a cornerstone of modern drug discovery, offering the ability to efficiently manage and analyze large volumes of chemical and biological data. Publicly available databases such as PubChem, ZINC, ChEMBL, DrugBank, ChemDiv, natural product databases, among others, are essential for accessing diverse chemical structures, biological activities, and pharmacological properties.
Areas covered: This review provides an overview of recent (2024-2025) trends in mining data from PubChem and other representative public databases for virtual screening. It also discusses the integration of experimental validation and computational tools in drug design and cheminformatics workflows. The article is based on literature retrieved from SciFinder.
Expert opinion: Public chemical databases contain thousands to billions of compounds and various computational strategies have necessitated development to navigate this vast chemical space effectively. These include application programming interfaces, similarity searches, physicochemical filtering, and target-based selection. Such filtering strategies have enabled the extraction of focused compound subsets for evaluation through various cheminformatics tools, ultimately supporting informed decision-making in lead discovery and optimization.
{"title":"Exploiting PubChem and other public databases for virtual screening in 2025: what are the latest trends?","authors":"Alberto Marbán-González, Verónica Ramírez-Cid, Alejandro Cristóbal-Ramírez, José L Medina-Franco","doi":"10.1080/17460441.2025.2558161","DOIUrl":"10.1080/17460441.2025.2558161","url":null,"abstract":"<p><strong>Introduction: </strong>Cheminformatics has become a cornerstone of modern drug discovery, offering the ability to efficiently manage and analyze large volumes of chemical and biological data. Publicly available databases such as PubChem, ZINC, ChEMBL, DrugBank, ChemDiv, natural product databases, among others, are essential for accessing diverse chemical structures, biological activities, and pharmacological properties.</p><p><strong>Areas covered: </strong>This review provides an overview of recent (2024-2025) trends in mining data from PubChem and other representative public databases for virtual screening. It also discusses the integration of experimental validation and computational tools in drug design and cheminformatics workflows. The article is based on literature retrieved from SciFinder.</p><p><strong>Expert opinion: </strong>Public chemical databases contain thousands to billions of compounds and various computational strategies have necessitated development to navigate this vast chemical space effectively. These include application programming interfaces, similarity searches, physicochemical filtering, and target-based selection. Such filtering strategies have enabled the extraction of focused compound subsets for evaluation through various cheminformatics tools, ultimately supporting informed decision-making in lead discovery and optimization.</p>","PeriodicalId":12267,"journal":{"name":"Expert Opinion on Drug Discovery","volume":" ","pages":"1387-1403"},"PeriodicalIF":4.9,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145000017","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}