首页 > 最新文献

Bioinformatics (Oxford, England)最新文献

英文 中文
MMPCS: multi-view molecular pretraining based on consistency information and specific information. MMPCS:基于一致性信息和特定信息的多视图分子预训练。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag028
Chenyang Xie, Yingying Song, Song He, Xiaochen Bo, Zhongnan Zhang

Motivation: The goal of molecular representation learning is to automate the extraction of molecular features, a critical task in cheminformatics and drug discovery. While pretraining models using multiple views like SMILES, two-dimensional graphs, and three-dimensional conformations have advanced the field, integrating them effectively to produce superior representations remains a challenge.

Results: bridge this gap, we propose a novel multi-view molecular pretraining method termed MMPCS, which explicitly factorizes representations into consistency and specific information. Our approach utilizes the Graph Isomorphism Network and the RoBERTa model to encode two-dimensional molecular topological graphs and SMILES sequences, respectively. Each resulting molecular embedding is decomposed into a shared consistency component and a view-specific remainder. An autoencoder then aligns the consistency information across views. The combined consistency and view-specific representations serve as input for downstream tasks, enabling precise and task-aware predictions. When benchmarked against 16 state-of-the-art molecular pretraining methods, MMPCS achieved the highest average performance across both classification and regression tasks for molecular property prediction. It also delivered outstanding results in predicting drug-target binding affinity and cancer drug response, demonstrating its robustness and broad applicability. Additionally, a case study on the SARS-CoV-2 Omicron variant highlights the potential of MMPCS in facilitating drug repurposing efforts.

Availability and implementation: The source code and datasets supporting this study are publicly available at GitHub (https://github.com/xmubiocode/MMPCS) and Zenodo (https://doi.org/10.5281/zenodo.18182748).

动机:分子表征学习的目标是自动提取分子特征,这是化学信息学和药物发现的关键任务。虽然使用多种视图(如SMILES、二维图和三维构象)的预训练模型已经推动了该领域的发展,但有效地整合它们以产生更好的表示仍然是一个挑战。结果:为了弥补这一差距,我们提出了一种新的多视图分子预训练方法,称为MMPCS,它明确地将表征分解为一致性和特定信息。该方法利用图同构网络和RoBERTa模型分别对二维分子拓扑图和SMILES序列进行编码。每个结果的分子嵌入被分解成一个共享的一致性组件和一个特定于视图的余项。然后,自动编码器在视图之间对齐一致性信息。组合的一致性和特定于视图的表示作为下游任务的输入,支持精确和任务感知的预测。当与16种最先进的分子预训练方法进行基准测试时,MMPCS在分子性质预测的分类和回归任务中都取得了最高的平均性能。该方法在预测药物靶点结合亲和力和癌症药物反应方面也取得了出色的结果,证明了其稳健性和广泛的适用性。此外,对SARS-CoV-2 Omicron变体的案例研究强调了MMPCS在促进药物再利用方面的潜力。可用性和实现:支持本研究的源代码和数据集可在GitHub (https://github.com/xmubiocode/MMPCS)和Zenodo (https://doi.org/10.5281/zenodo.18182748)上公开获取。
{"title":"MMPCS: multi-view molecular pretraining based on consistency information and specific information.","authors":"Chenyang Xie, Yingying Song, Song He, Xiaochen Bo, Zhongnan Zhang","doi":"10.1093/bioinformatics/btag028","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag028","url":null,"abstract":"<p><strong>Motivation: </strong>The goal of molecular representation learning is to automate the extraction of molecular features, a critical task in cheminformatics and drug discovery. While pretraining models using multiple views like SMILES, two-dimensional graphs, and three-dimensional conformations have advanced the field, integrating them effectively to produce superior representations remains a challenge.</p><p><strong>Results: </strong>bridge this gap, we propose a novel multi-view molecular pretraining method termed MMPCS, which explicitly factorizes representations into consistency and specific information. Our approach utilizes the Graph Isomorphism Network and the RoBERTa model to encode two-dimensional molecular topological graphs and SMILES sequences, respectively. Each resulting molecular embedding is decomposed into a shared consistency component and a view-specific remainder. An autoencoder then aligns the consistency information across views. The combined consistency and view-specific representations serve as input for downstream tasks, enabling precise and task-aware predictions. When benchmarked against 16 state-of-the-art molecular pretraining methods, MMPCS achieved the highest average performance across both classification and regression tasks for molecular property prediction. It also delivered outstanding results in predicting drug-target binding affinity and cancer drug response, demonstrating its robustness and broad applicability. Additionally, a case study on the SARS-CoV-2 Omicron variant highlights the potential of MMPCS in facilitating drug repurposing efforts.</p><p><strong>Availability and implementation: </strong>The source code and datasets supporting this study are publicly available at GitHub (https://github.com/xmubiocode/MMPCS) and Zenodo (https://doi.org/10.5281/zenodo.18182748).</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145986087","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
Semantic-Enhanced heterogeneous graph learning for identifying ncRNAs associated with drug resistance. 语义增强异构图学习识别与耐药相关的ncrna。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag029
Hang Wei, Yuran Xie, Wenxiang Zhang, Linyang Li, Shuai Wu, Lin Gao

Motivation: Identifying non-coding RNAs (ncRNAs) associated with drug resistance is critical for elucidating molecular mechanisms underlying drug response, facilitating drug screening, and discovering novel therapeutic targets. While several graph neural network-based methods have been proposed to infer ncRNA-drug resistance associations, they remain fundamentally constrained by semantic distortion induced by sparse bipartite network and neglect of relational semantics among molecular entities, ultimately compromising both predictive reliability and biological interpretability.

Results: In this study, we propose iNcRD-HG, a novel framework for identifying ncRNA-drug resistance associations. The framework addresses three critical aspects: constructing a context-enriched heterogeneous network that integrates six distinct molecular interaction types with bio-entity-specific attributes, developing a semantic-enhanced graph learning architecture that implements relation-type-aware message passing to capture complex contextual dependencies, and introducing an interpretability mechanism to reveal potential synergistic pathways underlying drug response. Experimental results demonstrate that iNcRD-HG achieves superior predictive performance across diverse benchmark datasets while deriving association features with strong discriminative capability. By identifying molecular synergistic contexts, iNcRD-HG provides mechanistically interpretable insights into ncRNA-mediated drug resistance.

Availability and implementation: Datasets and source codes are available at https://github.com/Biohang/iNcRD-HG.

Supplementary information: Supplementary data are available at Bioinformatics online.

动机:鉴定与耐药相关的非编码rna (ncRNAs)对于阐明药物反应的分子机制、促进药物筛选和发现新的治疗靶点至关重要。虽然已经提出了几种基于图神经网络的方法来推断ncrna -耐药关联,但它们仍然受到稀疏二部网络引起的语义扭曲和忽视分子实体之间的关系语义的限制,最终损害了预测可靠性和生物学可解释性。结果:在这项研究中,我们提出了一个新的框架iNcRD-HG,用于鉴定ncrna -耐药关联。该框架涉及三个关键方面:构建一个上下文丰富的异构网络,该网络集成了六种具有生物实体特定属性的不同分子相互作用类型;开发一个语义增强的图学习架构,实现关系类型感知的消息传递,以捕获复杂的上下文依赖性;引入可解释性机制,以揭示药物反应背后的潜在协同途径。实验结果表明,iNcRD-HG在不同的基准数据集上取得了优异的预测性能,同时获得了具有较强判别能力的关联特征。通过识别分子协同背景,iNcRD-HG为ncrna介导的耐药提供了机制解释。可用性和实施:数据集和源代码可在https://github.com/Biohang/iNcRD-HG.Supplementary信息上获得;补充数据可在Bioinformatics在线上获得。
{"title":"Semantic-Enhanced heterogeneous graph learning for identifying ncRNAs associated with drug resistance.","authors":"Hang Wei, Yuran Xie, Wenxiang Zhang, Linyang Li, Shuai Wu, Lin Gao","doi":"10.1093/bioinformatics/btag029","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag029","url":null,"abstract":"<p><strong>Motivation: </strong>Identifying non-coding RNAs (ncRNAs) associated with drug resistance is critical for elucidating molecular mechanisms underlying drug response, facilitating drug screening, and discovering novel therapeutic targets. While several graph neural network-based methods have been proposed to infer ncRNA-drug resistance associations, they remain fundamentally constrained by semantic distortion induced by sparse bipartite network and neglect of relational semantics among molecular entities, ultimately compromising both predictive reliability and biological interpretability.</p><p><strong>Results: </strong>In this study, we propose iNcRD-HG, a novel framework for identifying ncRNA-drug resistance associations. The framework addresses three critical aspects: constructing a context-enriched heterogeneous network that integrates six distinct molecular interaction types with bio-entity-specific attributes, developing a semantic-enhanced graph learning architecture that implements relation-type-aware message passing to capture complex contextual dependencies, and introducing an interpretability mechanism to reveal potential synergistic pathways underlying drug response. Experimental results demonstrate that iNcRD-HG achieves superior predictive performance across diverse benchmark datasets while deriving association features with strong discriminative capability. By identifying molecular synergistic contexts, iNcRD-HG provides mechanistically interpretable insights into ncRNA-mediated drug resistance.</p><p><strong>Availability and implementation: </strong>Datasets and source codes are available at https://github.com/Biohang/iNcRD-HG.</p><p><strong>Supplementary information: </strong>Supplementary data are available at Bioinformatics online.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145986071","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
CADS: A Causal Inference Framework for Identifying Essential Genes to Enhance Drug Synergy Prediction. CADS:鉴定必要基因以增强药物协同作用预测的因果推理框架。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag010
Huaiwu Zhang, Xinliang Sun, Jianxin Wang, Min Li, Jing Tang

Motivation: Drug synergy is crucial for developing effective combination therapies, but traditional screening methods suffer from inefficiency and high costs. While deep learning shows promise for predicting drug synergy, current approaches using Transformers and graph neural networks focus on combining drug and cell line features without modelling how genes causally influence drug responses.

Results: To address this limitation, we propose CADS (Causal Adjustment for Drug Synergy), a deep learning framework that integrates causal relationships between genes and drug responses. Leveraging multi-omics data, CADS uses a learnable mask mechanism to identify key causal genes while filtering out irrelevant genetic factors through backdoor adjustment. Our model achieves two key objectives simultaneously: accurate prediction of drug synergy and interpretable causal gene discovery. Experiments on multiple datasets show that CADS consistently outperforms state-of-the-art methods across multiple metrics. Case studies demonstrate that CADS can reduce unnecessary complexity while providing more biological insights through its gene importance scores, which help identify clinically validated cancer-related genes that mediate drug interactions.

Availability and implementation: Taken together, CADS advances combination therapy prediction by explicitly modelling drug synergy causal genes, offering enhanced interpretability for AI-based drug development. The source code can be found at https://github.com/HuaiwuZhang/causalDC.

Supplementary information: Supplementary data are available at Bioinformatics online.

动机:药物协同作用对于开发有效的联合疗法至关重要,但传统的筛选方法效率低下且成本高。虽然深度学习有望预测药物协同作用,但目前使用transformer和图神经网络的方法侧重于结合药物和细胞系特征,而没有模拟基因如何因果影响药物反应。为了解决这一限制,我们提出了CADS(因果调整药物协同),这是一个深度学习框架,整合了基因和药物反应之间的因果关系。CADS利用多组学数据,利用可学习的掩模机制识别关键的致病基因,同时通过后门调节过滤掉无关的遗传因素。我们的模型同时实现了两个关键目标:准确预测药物协同作用和发现可解释的因果基因。在多个数据集上的实验表明,CADS在多个指标上始终优于最先进的方法。案例研究表明,CADS可以减少不必要的复杂性,同时通过其基因重要性评分提供更多的生物学见解,这有助于识别经临床验证的介导药物相互作用的癌症相关基因。可用性和实施:总的来说,CADS通过明确建模药物协同作用因果基因来推进联合治疗预测,为基于人工智能的药物开发提供增强的可解释性。源代码可在https://github.com/HuaiwuZhang/causalDC.Supplementary信息中找到:补充数据可在Bioinformatics在线获得。
{"title":"CADS: A Causal Inference Framework for Identifying Essential Genes to Enhance Drug Synergy Prediction.","authors":"Huaiwu Zhang, Xinliang Sun, Jianxin Wang, Min Li, Jing Tang","doi":"10.1093/bioinformatics/btag010","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag010","url":null,"abstract":"<p><strong>Motivation: </strong>Drug synergy is crucial for developing effective combination therapies, but traditional screening methods suffer from inefficiency and high costs. While deep learning shows promise for predicting drug synergy, current approaches using Transformers and graph neural networks focus on combining drug and cell line features without modelling how genes causally influence drug responses.</p><p><strong>Results: </strong>To address this limitation, we propose CADS (Causal Adjustment for Drug Synergy), a deep learning framework that integrates causal relationships between genes and drug responses. Leveraging multi-omics data, CADS uses a learnable mask mechanism to identify key causal genes while filtering out irrelevant genetic factors through backdoor adjustment. Our model achieves two key objectives simultaneously: accurate prediction of drug synergy and interpretable causal gene discovery. Experiments on multiple datasets show that CADS consistently outperforms state-of-the-art methods across multiple metrics. Case studies demonstrate that CADS can reduce unnecessary complexity while providing more biological insights through its gene importance scores, which help identify clinically validated cancer-related genes that mediate drug interactions.</p><p><strong>Availability and implementation: </strong>Taken together, CADS advances combination therapy prediction by explicitly modelling drug synergy causal genes, offering enhanced interpretability for AI-based drug development. The source code can be found at https://github.com/HuaiwuZhang/causalDC.</p><p><strong>Supplementary information: </strong>Supplementary data are available at Bioinformatics online.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971677","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
CMAtlas: a comprehensive DNA methylation atlas for exploring epigenetic alterations in 34 human cancer types. CMAtlas:一个全面的DNA甲基化图谱,用于探索34种人类癌症类型的表观遗传改变。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag022
Mengni Liu, Lizhen Jiang, Luowanyue Zhang, Tianjian Chen, Xingzhe Wang, Yuan Liang, Xianping Shi, Jian Ren, Yueyuan Zheng

Motivation: Aberrant DNA methylation is a fundamental epigenetic hallmark of cancer. However, existing resources often lack technological diversity and comprehensive cancer coverage. Furthermore, most platforms fail to achieve deep multi-omics integration and tend to ignore cancer-type-specific methylation features, limiting their utility in precision oncology and drug discovery.

Results: We developed CMAtlas (Cancer Methylation Atlas), a comprehensive platform integrating 13,753 samples across 34 cancer types. By applying technology-tailored pipelines to data from various profiling technologies, we identified 830,725 tumor-specific differentially methylated elements (DMEs) and 1,480,098 differentially methylated regions (DMRs), alongside 1,154,256 cancer-type-specific DMEs and 329,154 DMRs. The platform demonstrates high cross-platform consistency and strong concordance between tumor tissues and cell lines, ensuring the robustness of our findings. All DMEs and DMRs are annotated with multi-omics data (RNA expression, somatic mutations, and chromatin accessibility) and clinical relevance (survival associations and cell-free DNA profiling). We further demonstrate the utility of CMAtlas by identifying prognostic aberrant methylation in colorectal cancer driver genes.

Availability: CMAtlas is freely accessible at {{https://cmatlas.renlab.cn/}}. The platform offers an intuitive web interface supporting gene-centric and cancer-centric queries, alongside customizable analysis modules designed to facilitate user-specific research needs.

Supplementary information: Supplementary data are available at Bioinformatics online.

动机:异常DNA甲基化是癌症的基本表观遗传标志。然而,现有资源往往缺乏技术多样性和全面的癌症覆盖。此外,大多数平台未能实现深度多组学整合,并倾向于忽略癌症类型特异性甲基化特征,限制了它们在精确肿瘤学和药物发现中的应用。结果:我们开发了CMAtlas(癌症甲基化图谱),这是一个综合平台,整合了34种癌症类型的13,753个样本。通过将技术定制的管道应用于各种分析技术的数据,我们确定了830,725个肿瘤特异性差异甲基化元件(DMEs)和1,480,098个差异甲基化区域(DMRs),以及1,154,256个癌症类型特异性DMEs和329,154个DMRs。该平台具有高度的跨平台一致性和肿瘤组织和细胞系之间的强一致性,确保了我们研究结果的稳健性。所有DMEs和DMRs都用多组学数据(RNA表达、体细胞突变和染色质可及性)和临床相关性(生存关联和无细胞DNA分析)进行了注释。我们通过鉴定结直肠癌驱动基因的预后异常甲基化进一步证明了CMAtlas的效用。可用性:CMAtlas可在{{https://cmatlas.renlab.cn/}}免费访问。该平台提供了一个直观的网络界面,支持以基因为中心和以癌症为中心的查询,以及可定制的分析模块,旨在促进用户特定的研究需求。补充信息:补充数据可在生物信息学在线获取。
{"title":"CMAtlas: a comprehensive DNA methylation atlas for exploring epigenetic alterations in 34 human cancer types.","authors":"Mengni Liu, Lizhen Jiang, Luowanyue Zhang, Tianjian Chen, Xingzhe Wang, Yuan Liang, Xianping Shi, Jian Ren, Yueyuan Zheng","doi":"10.1093/bioinformatics/btag022","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag022","url":null,"abstract":"<p><strong>Motivation: </strong>Aberrant DNA methylation is a fundamental epigenetic hallmark of cancer. However, existing resources often lack technological diversity and comprehensive cancer coverage. Furthermore, most platforms fail to achieve deep multi-omics integration and tend to ignore cancer-type-specific methylation features, limiting their utility in precision oncology and drug discovery.</p><p><strong>Results: </strong>We developed CMAtlas (Cancer Methylation Atlas), a comprehensive platform integrating 13,753 samples across 34 cancer types. By applying technology-tailored pipelines to data from various profiling technologies, we identified 830,725 tumor-specific differentially methylated elements (DMEs) and 1,480,098 differentially methylated regions (DMRs), alongside 1,154,256 cancer-type-specific DMEs and 329,154 DMRs. The platform demonstrates high cross-platform consistency and strong concordance between tumor tissues and cell lines, ensuring the robustness of our findings. All DMEs and DMRs are annotated with multi-omics data (RNA expression, somatic mutations, and chromatin accessibility) and clinical relevance (survival associations and cell-free DNA profiling). We further demonstrate the utility of CMAtlas by identifying prognostic aberrant methylation in colorectal cancer driver genes.</p><p><strong>Availability: </strong>CMAtlas is freely accessible at {{https://cmatlas.renlab.cn/}}. The platform offers an intuitive web interface supporting gene-centric and cancer-centric queries, alongside customizable analysis modules designed to facilitate user-specific research needs.</p><p><strong>Supplementary information: </strong>Supplementary data are available at Bioinformatics online.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145986016","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
Characterizing Clinical Toxicity in Cancer Combination Therapies. 肿瘤联合治疗的临床毒性特征。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag007
Alexandra M Wong, Cecile Meier-Scherling, Lorin Crawford

Motivation: Predicting synergistic cancer drug combinations through computational methods offers a scalable approach to creating therapies that are more effective and less toxic. However, most algorithms focus solely on synergy without considering toxicity when selecting optimal drug combinations. In the absence of combinatorial toxicity assays, a few models use toxicity penalties to balance high synergy with lower toxicity. Still, these penalties have not been explicitly validated against known drug-drug interactions.

Results: In this study, we examine whether synergy scores and toxicity metrics correlate with known adverse drug interactions. While some metrics show trends with toxicity levels, our results reveal significant limitations in using them as penalties. These findings highlight the challenges of incorporating toxicity into synergy prediction frameworks and suggest that advancing the field requires more comprehensive combination toxicity data.

Availability and implementation: The code written for this project is available at https://github.com/amw14/toxicity-cancer-drug-combination.

动机:通过计算方法预测协同抗癌药物组合为创造更有效、毒性更小的治疗方法提供了一种可扩展的方法。然而,在选择最佳药物组合时,大多数算法只关注协同作用而不考虑毒性。在没有组合毒性试验的情况下,一些模型使用毒性惩罚来平衡高协同作用和低毒性。尽管如此,这些惩罚还没有明确地针对已知的药物-药物相互作用进行验证。结果:在本研究中,我们研究了协同作用评分和毒性指标是否与已知的不良药物相互作用相关。虽然一些指标显示了毒性水平的趋势,但我们的研究结果表明,使用它们作为惩罚措施存在重大局限性。这些发现突出了将毒性纳入协同作用预测框架的挑战,并表明推进该领域需要更全面的联合毒性数据。可用性和实现:为这个项目编写的代码可在https://github.com/amw14/toxicity-cancer-drug-combination上获得。
{"title":"Characterizing Clinical Toxicity in Cancer Combination Therapies.","authors":"Alexandra M Wong, Cecile Meier-Scherling, Lorin Crawford","doi":"10.1093/bioinformatics/btag007","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag007","url":null,"abstract":"<p><strong>Motivation: </strong>Predicting synergistic cancer drug combinations through computational methods offers a scalable approach to creating therapies that are more effective and less toxic. However, most algorithms focus solely on synergy without considering toxicity when selecting optimal drug combinations. In the absence of combinatorial toxicity assays, a few models use toxicity penalties to balance high synergy with lower toxicity. Still, these penalties have not been explicitly validated against known drug-drug interactions.</p><p><strong>Results: </strong>In this study, we examine whether synergy scores and toxicity metrics correlate with known adverse drug interactions. While some metrics show trends with toxicity levels, our results reveal significant limitations in using them as penalties. These findings highlight the challenges of incorporating toxicity into synergy prediction frameworks and suggest that advancing the field requires more comprehensive combination toxicity data.</p><p><strong>Availability and implementation: </strong>The code written for this project is available at https://github.com/amw14/toxicity-cancer-drug-combination.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971697","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
A Case-Based Explainable Graph Neural Network Framework for Mechanistic Drug Repositioning. 一种基于案例的可解释图神经网络框架用于机械药物重新定位。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag008
Adriana Carolina Gonzalez-Cavazos, Roger Tu, Meghamala Sinha, Andrew I Su

Drug repositioning offers a cost-effective alternative to traditional drug development by identifying new uses for existing drugs. Recent advances leverage Graph Neural Networks (GNN) to model complex biological data, showing promise in predicting novel drug-disease associations. However, these frameworks often lack explainability, a critical factor for validating predictions and understanding drug mechanisms. Here, we introduce Drug-Based Reasoning Explainer (DBR-X), an explainable GNN model that combines a link prediction module and a path-identification module to generate interpretable and faithful explanations. When benchmarked against other GNN link prediction frameworks, DBR-X achieves superior performance in identifying known drug-disease associations, demonstrating higher accuracy across all evaluation metrics. The quality of DBR-X biological explanations was assessed through multiple approaches: comparison with manually-curated drug mechanisms, evaluation of explanation faithfulness through deletion and insertion studies, and measurement of stability under graph perturbations. Together, our model not only advances the state-of-the-art in drug repositioning predictions but also provides multi-hop explanations that can accelerate the translation of computational predictions into clinical applications.

药物重新定位通过确定现有药物的新用途,为传统药物开发提供了一种具有成本效益的替代方案。最近的进展利用图神经网络(GNN)来模拟复杂的生物数据,在预测新的药物-疾病关联方面显示出希望。然而,这些框架往往缺乏可解释性,这是验证预测和理解药物机制的关键因素。本文介绍了基于药物的推理解释器(Drug-Based Reasoning Explainer, DBR-X),这是一种可解释的GNN模型,它结合了链接预测模块和路径识别模块,以生成可解释和可靠的解释。当与其他GNN链接预测框架进行基准比较时,DBR-X在识别已知药物-疾病关联方面表现优异,在所有评估指标中都显示出更高的准确性。通过多种方法评估DBR-X生物学解释的质量:与人工编制的药物机制进行比较,通过删除和插入研究评估解释的可信度,以及测量图扰动下的稳定性。总之,我们的模型不仅推进了最先进的药物重新定位预测,而且提供了多跳解释,可以加速将计算预测转化为临床应用。
{"title":"A Case-Based Explainable Graph Neural Network Framework for Mechanistic Drug Repositioning.","authors":"Adriana Carolina Gonzalez-Cavazos, Roger Tu, Meghamala Sinha, Andrew I Su","doi":"10.1093/bioinformatics/btag008","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag008","url":null,"abstract":"<p><p>Drug repositioning offers a cost-effective alternative to traditional drug development by identifying new uses for existing drugs. Recent advances leverage Graph Neural Networks (GNN) to model complex biological data, showing promise in predicting novel drug-disease associations. However, these frameworks often lack explainability, a critical factor for validating predictions and understanding drug mechanisms. Here, we introduce Drug-Based Reasoning Explainer (DBR-X), an explainable GNN model that combines a link prediction module and a path-identification module to generate interpretable and faithful explanations. When benchmarked against other GNN link prediction frameworks, DBR-X achieves superior performance in identifying known drug-disease associations, demonstrating higher accuracy across all evaluation metrics. The quality of DBR-X biological explanations was assessed through multiple approaches: comparison with manually-curated drug mechanisms, evaluation of explanation faithfulness through deletion and insertion studies, and measurement of stability under graph perturbations. Together, our model not only advances the state-of-the-art in drug repositioning predictions but also provides multi-hop explanations that can accelerate the translation of computational predictions into clinical applications.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145986038","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
GAMMA: Gap-aware Motif Mining under Incomplete Labeling with Applications to MHC Motifs. GAMMA:不完全标记下的间隙感知基序挖掘及其在MHC基序中的应用。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag014
Xinyi Tang, Ran Liu

Motivation: Sequence motif identification is crucial for understanding molecular recognition, particularly in immune responses involving peptide binding to MHC class I molecules for antigen presentation to T cells. Traditionally, MHC class I binding motifs are assumed to be contiguous and span nine amino acids. However, structural evidence suggests that binding may involve non-adjacent residues, challenging the assumptions of existing methods.

Results: In this study, we propose GAMMA (Gap-Aware Motif Mining Algorithm), a probabilistic framework designed to identify non-contiguous motifs under conditions of incomplete labeling. GAMMA employs Bayesian inference with MCMC sampling to jointly estimate motif parameters, binding locations, and the relative spacing between binding positions. Through extensive simulations and real-world applications to MHC class I peptide datasets, GAMMA outperforms existing motif discovery tools such as GLAM2 in accurately localizing binding residues and identifying the underlying motifs. Notably, our results suggest that the true number of binding residues may be eight, fewer than the commonly assumed nine. In addition, for longer peptides, the model captures increased flexibility in the central region, consistent with structural observations that peptides may bulge in the middle.

Availability: The raw data and the source codes are available on GitHub (https://github.com/RanLIUaca/GAMMAmotif).

Supplementary information: Supplementary data are available at Bioinformatics online.

动机:序列基序识别对于理解分子识别至关重要,特别是在涉及肽与MHC I类分子结合以向T细胞呈递抗原的免疫反应中。传统上,MHC I类结合基序被认为是连续的,跨越9个氨基酸。然而,结构证据表明,结合可能涉及非相邻残基,挑战现有方法的假设。结果:在本研究中,我们提出了GAMMA (Gap-Aware Motif Mining Algorithm),这是一个概率框架,旨在识别不完全标记条件下的非连续Motif。GAMMA使用贝叶斯推理和MCMC采样来联合估计基序参数、结合位置和结合位置之间的相对间距。通过对MHC I类肽数据集的广泛模拟和实际应用,GAMMA在精确定位结合残基和识别潜在基序方面优于现有的基序发现工具,如GLAM2。值得注意的是,我们的结果表明,结合残基的真实数量可能是8个,少于通常假设的9个。此外,对于较长的肽,该模型捕获了中心区域增加的灵活性,这与结构观察结果一致,即肽可能在中间凸起。可用性:原始数据和源代码可在GitHub上获得(https://github.com/RanLIUaca/GAMMAmotif).Supplementary information:补充数据可在Bioinformatics在线获得。
{"title":"GAMMA: Gap-aware Motif Mining under Incomplete Labeling with Applications to MHC Motifs.","authors":"Xinyi Tang, Ran Liu","doi":"10.1093/bioinformatics/btag014","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag014","url":null,"abstract":"<p><strong>Motivation: </strong>Sequence motif identification is crucial for understanding molecular recognition, particularly in immune responses involving peptide binding to MHC class I molecules for antigen presentation to T cells. Traditionally, MHC class I binding motifs are assumed to be contiguous and span nine amino acids. However, structural evidence suggests that binding may involve non-adjacent residues, challenging the assumptions of existing methods.</p><p><strong>Results: </strong>In this study, we propose GAMMA (Gap-Aware Motif Mining Algorithm), a probabilistic framework designed to identify non-contiguous motifs under conditions of incomplete labeling. GAMMA employs Bayesian inference with MCMC sampling to jointly estimate motif parameters, binding locations, and the relative spacing between binding positions. Through extensive simulations and real-world applications to MHC class I peptide datasets, GAMMA outperforms existing motif discovery tools such as GLAM2 in accurately localizing binding residues and identifying the underlying motifs. Notably, our results suggest that the true number of binding residues may be eight, fewer than the commonly assumed nine. In addition, for longer peptides, the model captures increased flexibility in the central region, consistent with structural observations that peptides may bulge in the middle.</p><p><strong>Availability: </strong>The raw data and the source codes are available on GitHub (https://github.com/RanLIUaca/GAMMAmotif).</p><p><strong>Supplementary information: </strong>Supplementary data are available at Bioinformatics online.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971709","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
Improving biomedical entity linking with generative relevance feedback. 利用生成相关性反馈改进生物医学实体链接。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag011
Darya Shlyk, Lawrence Hunter

Motivation: Biomedical Entity Linking (BEL) maps mentions in biomedical text to standardized identifiers, enabling structured data integration and downstream knowledge discovery. However, current BEL systems remain fundamentally constrained by the recall of the initial candidate pool, where suboptimal retrieval limits the overall effectiveness of the normalization pipeline.

Results: We present the first systematic evaluation of Generative Relevance Feedback (GRF) for enhancing candidate retrieval in state-of-the-art BEL systems. GRF leverages large language models (LLMs) to enrich the expressiveness of the mention in a zero-shot fashion. We assess GRF's impact under two scenarios-direct linking prediction and candidate generation in cascading normalization pipelines-and analyze its sensitivity to different LLMs, feedback types, and integration strategies. Experiments across eight corpora and four biomedical knowledge bases demonstrate that integrating GRF significantly improves both accuracy and recall, thereby increasing the upper bound on normalization performance. Our findings highlight GRF as an efficient, model-agnostic solution and underscore its potential as a key component for advancing BEL.

Availability: The code to reproduce our experiments can be found at: https://doi.org/10.5281/zenodo.17853541.

动机:生物医学实体链接(BEL)将生物医学文本中的提及映射到标准化标识符,从而实现结构化数据集成和下游知识发现。然而,当前的BEL系统仍然从根本上受到初始候选池召回的限制,其中次优检索限制了规范化管道的整体有效性。结果:我们首次对生成关联反馈(GRF)进行了系统评估,以增强最先进的BEL系统中的候选检索。GRF利用大型语言模型(llm)以零射击的方式丰富提及的表达性。我们评估了GRF在级联归一化管道中直接链接预测和候选生成两种情况下的影响,并分析了其对不同llm、反馈类型和集成策略的敏感性。基于8个语料库和4个生物医学知识库的实验表明,整合GRF显著提高了正确率和召回率,从而提高了归一化性能的上限。我们的发现强调了GRF是一种高效的、与模型无关的解决方案,并强调了它作为推进bel的关键组件的潜力。可用性:可以在https://doi.org/10.5281/zenodo.17853541上找到重现我们实验的代码。
{"title":"Improving biomedical entity linking with generative relevance feedback.","authors":"Darya Shlyk, Lawrence Hunter","doi":"10.1093/bioinformatics/btag011","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag011","url":null,"abstract":"<p><strong>Motivation: </strong>Biomedical Entity Linking (BEL) maps mentions in biomedical text to standardized identifiers, enabling structured data integration and downstream knowledge discovery. However, current BEL systems remain fundamentally constrained by the recall of the initial candidate pool, where suboptimal retrieval limits the overall effectiveness of the normalization pipeline.</p><p><strong>Results: </strong>We present the first systematic evaluation of Generative Relevance Feedback (GRF) for enhancing candidate retrieval in state-of-the-art BEL systems. GRF leverages large language models (LLMs) to enrich the expressiveness of the mention in a zero-shot fashion. We assess GRF's impact under two scenarios-direct linking prediction and candidate generation in cascading normalization pipelines-and analyze its sensitivity to different LLMs, feedback types, and integration strategies. Experiments across eight corpora and four biomedical knowledge bases demonstrate that integrating GRF significantly improves both accuracy and recall, thereby increasing the upper bound on normalization performance. Our findings highlight GRF as an efficient, model-agnostic solution and underscore its potential as a key component for advancing BEL.</p><p><strong>Availability: </strong>The code to reproduce our experiments can be found at: https://doi.org/10.5281/zenodo.17853541.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971651","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
scSNViz: Visualization and analysis of Cell-Specific expressed SNVs. scSNViz:细胞特异性表达snv的可视化和分析。
IF 5.4 Pub Date : 2026-01-14 DOI: 10.1093/bioinformatics/btag023
Siera Martinez, Tushar Sharma, Luke Johnson, Allen Kim, Vania Ballesteros Prieto, Hovhannes Arestakesyan, Sunisha Harish, Jewel Dias, Joseph Goldfrank, Nathan Edwards, Anelia Horvath

Motivation: Accurately characterizing expressed genetic variation at the single-cell level is essential for understanding transcriptional heterogeneity, allelic regulation, and mutational dynamics within complex tissues. However, few tools enable comprehensive visualization and quantitative analysis of expressed variants across individual cells.

Results: scSNViz is an R package for the exploration, quantification, and visualization of expressed single-nucleotide variants (SNVs) from cell-barcoded single-cell RNA sequencing (scRNA-seq) data. The software supports estimation of variant allele fractions, clustering of SNV expression profiles, and 2D and 3D visualization of individual SNVs or user-defined SNV groups. Beyond visualization, scSNViz facilitates investigation of cell-, cluster-, or lineage-specific variant expression patterns, as well as allelic dynamics including imprinting, random allele inactivation, and transcriptional bursting. It interoperates seamlessly with established single-cell frameworks-Seurat for clustering, Slingshot for trajectory inference, scType for cell-type annotation, and CopyKat for copy-number profiling-enabling integrative multi-omic analyses of expressed variation.

Availability: scSNViz is implemented in R and freely available at https://github.com/HorvathLab/scSNViz (DOI: 10.5281/zenodo.17307516). The package includes comprehensive documentation and example workflows designed for users with limited bioinformatics experience.

Supplementary information: Supplementary data are available at Bioinformatics online.

动机:准确地描述单细胞水平上表达的遗传变异对于理解复杂组织中的转录异质性、等位基因调控和突变动力学至关重要。然而,很少有工具能够对单个细胞的表达变异进行全面的可视化和定量分析。结果:scSNViz是一个R软件包,用于从细胞条形码单细胞RNA测序(scRNA-seq)数据中探索、量化和可视化表达的单核苷酸变异(snv)。该软件支持变异等位基因分数的估计,SNV表达谱的聚类,以及单个SNV或用户定义的SNV组的2D和3D可视化。除了可视化之外,scSNViz还有助于研究细胞、集群或谱系特异性变异表达模式,以及等位基因动力学,包括印迹、随机等位基因失活和转录破裂。它与已建立的单细胞框架(seurat用于聚类,Slingshot用于轨迹推断,scType用于细胞类型注释,CopyKat用于拷贝数分析)无缝互操作,从而实现表达变异的综合多组学分析。可用性:scSNViz是用R实现的,可以在https://github.com/HorvathLab/scSNViz免费获得(DOI: 10.5281/zenodo.17307516)。该软件包包括全面的文档和示例工作流程,为有限的生物信息学经验的用户设计。补充信息:补充数据可在生物信息学在线获取。
{"title":"scSNViz: Visualization and analysis of Cell-Specific expressed SNVs.","authors":"Siera Martinez, Tushar Sharma, Luke Johnson, Allen Kim, Vania Ballesteros Prieto, Hovhannes Arestakesyan, Sunisha Harish, Jewel Dias, Joseph Goldfrank, Nathan Edwards, Anelia Horvath","doi":"10.1093/bioinformatics/btag023","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag023","url":null,"abstract":"<p><strong>Motivation: </strong>Accurately characterizing expressed genetic variation at the single-cell level is essential for understanding transcriptional heterogeneity, allelic regulation, and mutational dynamics within complex tissues. However, few tools enable comprehensive visualization and quantitative analysis of expressed variants across individual cells.</p><p><strong>Results: </strong>scSNViz is an R package for the exploration, quantification, and visualization of expressed single-nucleotide variants (SNVs) from cell-barcoded single-cell RNA sequencing (scRNA-seq) data. The software supports estimation of variant allele fractions, clustering of SNV expression profiles, and 2D and 3D visualization of individual SNVs or user-defined SNV groups. Beyond visualization, scSNViz facilitates investigation of cell-, cluster-, or lineage-specific variant expression patterns, as well as allelic dynamics including imprinting, random allele inactivation, and transcriptional bursting. It interoperates seamlessly with established single-cell frameworks-Seurat for clustering, Slingshot for trajectory inference, scType for cell-type annotation, and CopyKat for copy-number profiling-enabling integrative multi-omic analyses of expressed variation.</p><p><strong>Availability: </strong>scSNViz is implemented in R and freely available at https://github.com/HorvathLab/scSNViz (DOI: 10.5281/zenodo.17307516). The package includes comprehensive documentation and example workflows designed for users with limited bioinformatics experience.</p><p><strong>Supplementary information: </strong>Supplementary data are available at Bioinformatics online.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145986024","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
ChemGenXplore: An Interactive Tool for Exploring and Analysing Chemical Genomic Data. ChemGenXplore:一个用于探索和分析化学基因组数据的交互式工具。
IF 5.4 Pub Date : 2026-01-13 DOI: 10.1093/bioinformatics/btag021
Huda Ahmad, Hannah M Doherty, Sam Benedict, James Haycocks, Ge Zhou, Patrick Moynihan, Danesh Moradigaravand, Manuel Banzhaf

Motivation: Chemical genomics is a powerful high-throughput approach to systematically link phenotypes to genotypes. However, the vast datasets generated remain challenging to explore due to the lack of integrated, interactive tools for visualisation and analysis. Existing workflows often require multiple independent software tools, limiting data accessibility and collaboration. Therefore, we created a user-friendly platform that enables efficient exploration and sharing of chemical genomics data.

Results: We developed ChemGenXplore, a web-based Shiny application designed to streamline the visualisation and analysis of chemical genomic screens. It offers two primary functionalities: one for exploring pre-implemented datasets and another for analysing user-uploaded datasets. ChemGenXplore enables users to visualise phenotypic profiles, assess gene-gene and condition-condition correlations, perform GO and KEGG enrichment analysis, and generate customisable, interactive heatmaps. To further support collaborative research, ChemGenXplore also facilitates the comparative analysis of chemical genomic and other omics datasets. By consolidating these features into a single interactive and accessible tool, ChemGenXplore facilitates data sharing, enhances reproducibility, and promotes collaboration within the research community.

Availability: ChemGenXplore is freely accessible as a web application at https://chemgenxplore.kaust.edu.sa/. Source code and documentation, including instructions for local installation, are provided on GitHub (https://github.com/Hudaahmadd/ChemGenXplore). A Docker image is also available on DockerHub (https://hub.docker.com/r/hudaahmad/chemgenxplore) to ensure reproducibility and simplify installation.

Contact: example@example.org.

Supplementary information: Supplementary data are available at Bioinformatics online.

动机:化学基因组学是一种强大的高通量方法,可以系统地将表型与基因型联系起来。然而,由于缺乏集成的、交互式的可视化和分析工具,产生的大量数据集仍然具有挑战性。现有的工作流通常需要多个独立的软件工具,限制了数据的可访问性和协作。因此,我们创建了一个用户友好的平台,可以有效地探索和共享化学基因组学数据。结果:我们开发了ChemGenXplore,这是一个基于网络的Shiny应用程序,旨在简化化学基因组筛选的可视化和分析。它提供了两个主要功能:一个用于探索预实现的数据集,另一个用于分析用户上传的数据集。ChemGenXplore使用户能够可视化表型谱,评估基因-基因和条件-条件相关性,执行GO和KEGG富集分析,并生成可定制的交互式热图。为了进一步支持合作研究,ChemGenXplore还促进了化学基因组学和其他组学数据集的比较分析。ChemGenXplore将这些功能整合到一个单一的交互式和可访问的工具中,促进了数据共享,提高了可重复性,并促进了研究界的合作。可用性:ChemGenXplore作为web应用程序可免费访问https://chemgenxplore.kaust.edu.sa/。源代码和文档,包括本地安装的说明,在GitHub (https://github.com/Hudaahmadd/ChemGenXplore)上提供。DockerHub (https://hub.docker.com/r/hudaahmad/chemgenxplore)上也提供Docker镜像,以确保可再现性并简化安装。联系方式:example@example.org.Supplementary信息:补充数据可在Bioinformatics在线获取。
{"title":"ChemGenXplore: An Interactive Tool for Exploring and Analysing Chemical Genomic Data.","authors":"Huda Ahmad, Hannah M Doherty, Sam Benedict, James Haycocks, Ge Zhou, Patrick Moynihan, Danesh Moradigaravand, Manuel Banzhaf","doi":"10.1093/bioinformatics/btag021","DOIUrl":"https://doi.org/10.1093/bioinformatics/btag021","url":null,"abstract":"<p><strong>Motivation: </strong>Chemical genomics is a powerful high-throughput approach to systematically link phenotypes to genotypes. However, the vast datasets generated remain challenging to explore due to the lack of integrated, interactive tools for visualisation and analysis. Existing workflows often require multiple independent software tools, limiting data accessibility and collaboration. Therefore, we created a user-friendly platform that enables efficient exploration and sharing of chemical genomics data.</p><p><strong>Results: </strong>We developed ChemGenXplore, a web-based Shiny application designed to streamline the visualisation and analysis of chemical genomic screens. It offers two primary functionalities: one for exploring pre-implemented datasets and another for analysing user-uploaded datasets. ChemGenXplore enables users to visualise phenotypic profiles, assess gene-gene and condition-condition correlations, perform GO and KEGG enrichment analysis, and generate customisable, interactive heatmaps. To further support collaborative research, ChemGenXplore also facilitates the comparative analysis of chemical genomic and other omics datasets. By consolidating these features into a single interactive and accessible tool, ChemGenXplore facilitates data sharing, enhances reproducibility, and promotes collaboration within the research community.</p><p><strong>Availability: </strong>ChemGenXplore is freely accessible as a web application at https://chemgenxplore.kaust.edu.sa/. Source code and documentation, including instructions for local installation, are provided on GitHub (https://github.com/Hudaahmadd/ChemGenXplore). A Docker image is also available on DockerHub (https://hub.docker.com/r/hudaahmad/chemgenxplore) to ensure reproducibility and simplify installation.</p><p><strong>Contact: </strong>example@example.org.</p><p><strong>Supplementary information: </strong>Supplementary data are available at Bioinformatics online.</p>","PeriodicalId":93899,"journal":{"name":"Bioinformatics (Oxford, England)","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145967397","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
Bioinformatics (Oxford, England)
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:604180095
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