首页 > 最新文献

Advanced drug delivery reviews最新文献

英文 中文
Engineering tissue patterning in human stem cell-based embryo models 人类干细胞胚胎模型的工程组织模式
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-20 DOI: 10.1016/j.addr.2025.115765
Ella G. Lambert , Sara Romanazzo , Peter L.H. Newman , Kristopher A. Kilian
Human embryonic development is challenging to study in vitro as animal models inadequately represent human biology, while use of natural human embryos is both ethically and technically limited. Stem cell-based embryo models (SCBEMs) have emerged as a powerful alternative, enabling faithful recapitulation of early human development. However, current approaches predominantly rely on stochastic self-organisation with globally delivered signals, producing variable and often non-recapitulative structures. This review addresses this gap by introducing the first engineering-anchored taxonomy of human SCBEMs, systematically organizing the literature by their underlying technical platform rather than biological outcome alone. We demonstrate how five key engineering approaches – micropatterning, biomaterials, microwells, microfluidics, and dynamic culture – constrain morpho-and-histogenic patterning to determine developmental fidelity. We identify metabolic constraints limiting current models to ∼1 mm diameter as the primary bottleneck and demonstrate how vascular engineering and perfusion systems offer solutions. Finally, we propose standardisation metrics linking technical parameters to biological outcomes and establish an ethical framework defined by engineering choices.
人类胚胎发育的体外研究具有挑战性,因为动物模型不能充分代表人类生物学,而天然人类胚胎的使用在伦理和技术上都受到限制。基于干细胞的胚胎模型(SCBEMs)已经成为一种强大的替代方案,能够忠实地再现早期人类发育。然而,目前的方法主要依赖于具有全局传递信号的随机自组织,产生可变且通常是非概括的结构。本综述通过引入第一个以工程为基础的人类方案分类法来解决这一差距,系统地根据其潜在的技术平台而不是单独的生物学结果组织文献。我们展示了五种关键的工程方法——微模式、生物材料、微孔、微流体和动态培养——如何约束形态和组织结构模式来确定发育保真度。我们确定代谢限制将当前模型限制在1毫米直径为主要瓶颈,并展示血管工程和灌注系统如何提供解决方案。最后,我们提出了将技术参数与生物学结果联系起来的标准化指标,并建立了一个由工程选择定义的伦理框架。
{"title":"Engineering tissue patterning in human stem cell-based embryo models","authors":"Ella G. Lambert ,&nbsp;Sara Romanazzo ,&nbsp;Peter L.H. Newman ,&nbsp;Kristopher A. Kilian","doi":"10.1016/j.addr.2025.115765","DOIUrl":"10.1016/j.addr.2025.115765","url":null,"abstract":"<div><div>Human embryonic development is challenging to study in vitro as animal models inadequately represent human biology, while use of natural human embryos is both ethically and technically limited. Stem cell-based embryo models (SCBEMs) have emerged as a powerful alternative, enabling faithful recapitulation of early human development. However, current approaches predominantly rely on stochastic self-organisation with globally delivered signals, producing variable and often non-recapitulative structures. This review addresses this gap by introducing the first engineering-anchored taxonomy of human SCBEMs, systematically organizing the literature by their underlying technical platform rather than biological outcome alone. We demonstrate how five key engineering approaches – micropatterning, biomaterials, microwells, microfluidics, and dynamic culture – constrain morpho-and-histogenic patterning to determine developmental fidelity. We identify metabolic constraints limiting current models to ∼1 mm diameter as the primary bottleneck and demonstrate how vascular engineering and perfusion systems offer solutions. Finally, we propose standardisation metrics linking technical parameters to biological outcomes and establish an ethical framework defined by engineering choices.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115765"},"PeriodicalIF":17.6,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145784473","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Small data, big challenges: Machine- and deep-learning strategies for data-limited drug discovery 小数据,大挑战:机器和深度学习策略用于数据有限的药物发现
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-18 DOI: 10.1016/j.addr.2025.115762
Nazreen Pallikkavaliyaveetil , Sriram Chandrasekaran
A critical bottleneck limiting the potential of Machine Learning (ML) and Deep Learning (DL) models within the drug discovery and development (DDD) pipeline is the scarcity of high-quality experimental data. Limited data is not an anomaly but an inherent characteristic of the DDD process. Significant financial costs, time, and confidentiality concerns limit the scale of available datasets. Applying standard ML and DL algorithms directly to these small datasets presents substantial challenges. Traditional ML models remain constrained by their dependence on handcrafted features and limited ability to capture complex biological relationships. In contrast, DL algorithms that assume data abundance are prone to overfitting and poor generalization when trained on small datasets. The small data problem thus represents a fundamental constraint that shapes the practical utility and trustworthiness of AI applications in DDD. While prior reviews have surveyed the broad landscape of AI and ML in drug discovery, a significant gap exists concerning the small data challenge across the DDD pipeline. Addressing this challenge requires adapting DL methods that typically assume data abundance, while also extending traditional ML approaches that, although well-suited to small data, remain limited in their representational capacity. This review addresses this gap by surveying key drug discovery tasks, highlighting the prevalence of limited data, and synthesizing both traditional ML methods and advanced DL strategies tailored to these contexts. By integrating methodological advances with task-specific applications, the review outlines current approaches and identifies opportunities for advancing robust, interpretable, and generalizable AI in drug discovery.
在药物发现和开发(DDD)管道中限制机器学习(ML)和深度学习(DL)模型潜力的一个关键瓶颈是缺乏高质量的实验数据。有限的数据不是异常,而是DDD过程的固有特征。重大的财务成本、时间和机密性问题限制了可用数据集的规模。将标准的ML和DL算法直接应用于这些小数据集具有实质性的挑战。传统的机器学习模型仍然受到依赖手工制作的特征和捕获复杂生物关系的有限能力的限制。相比之下,假设数据丰富的DL算法在小数据集上训练时容易过度拟合和泛化不良。因此,小数据问题代表了影响DDD中人工智能应用的实际效用和可信度的基本约束。虽然之前的评论已经调查了药物发现中人工智能和机器学习的广泛前景,但在DDD管道的小数据挑战方面存在显着差距。解决这一挑战需要适应通常假设数据丰富的DL方法,同时也扩展传统的ML方法,这些方法虽然非常适合小数据,但在表示能力方面仍然有限。这篇综述通过调查关键的药物发现任务,突出有限数据的普遍性,以及综合传统的机器学习方法和针对这些背景量身定制的高级深度学习策略来解决这一差距。通过将方法上的进步与特定任务的应用相结合,本综述概述了当前的方法,并确定了在药物发现中推进稳健、可解释和可推广的人工智能的机会。
{"title":"Small data, big challenges: Machine- and deep-learning strategies for data-limited drug discovery","authors":"Nazreen Pallikkavaliyaveetil ,&nbsp;Sriram Chandrasekaran","doi":"10.1016/j.addr.2025.115762","DOIUrl":"10.1016/j.addr.2025.115762","url":null,"abstract":"<div><div>A critical bottleneck limiting the potential of Machine Learning (ML) and Deep Learning (DL) models within the drug discovery and development (DDD) pipeline is the scarcity of high-quality experimental data. Limited data is not an anomaly but an inherent characteristic of the DDD process. Significant financial costs, time, and confidentiality concerns limit the scale of available datasets. Applying standard ML and DL algorithms directly to these small datasets presents substantial challenges. Traditional ML models remain constrained by their dependence on handcrafted features and limited ability to capture complex biological relationships. In contrast, DL algorithms that assume data abundance are prone to overfitting and poor generalization when trained on small datasets. The small data problem thus represents a fundamental constraint that shapes the practical utility and trustworthiness of AI applications in DDD. While prior reviews have surveyed the broad landscape of AI and ML in drug discovery, a significant gap exists concerning the small data challenge across the DDD pipeline. Addressing this challenge requires adapting DL methods that typically assume data abundance, while also extending traditional ML approaches that, although well-suited to small data, remain limited in their representational capacity. This review addresses this gap by surveying key drug discovery tasks, highlighting the prevalence of limited data, and synthesizing both traditional ML methods and advanced DL strategies tailored to these contexts. By integrating methodological advances with task-specific applications, the review outlines current approaches and identifies opportunities for advancing robust, interpretable, and generalizable AI in drug discovery.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115762"},"PeriodicalIF":17.6,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145771264","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Editorial: Novel ultrasound-based strategies for precision therapeutics and visualization. 编辑:新的基于超声的精确治疗和可视化策略。
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-16 DOI: 10.1016/j.addr.2025.115763
Xinwu Cui, Xiaoyuan Chen
{"title":"Editorial: Novel ultrasound-based strategies for precision therapeutics and visualization.","authors":"Xinwu Cui, Xiaoyuan Chen","doi":"10.1016/j.addr.2025.115763","DOIUrl":"10.1016/j.addr.2025.115763","url":null,"abstract":"","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":" ","pages":"115763"},"PeriodicalIF":17.6,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145779775","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The growing impact of machine learning on drug formulation science 机器学习对药物配方科学的影响越来越大
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-12 DOI: 10.1016/j.addr.2025.115761
Zeqing Bao , Steven Huynh , Frantz Le Devedec , Jo Nguyen , Christine Allen
Machine learning (ML) has increasingly been adopted in drug formulation science to support more efficient, data-driven drug development strategies. A growing number of studies have demonstrated the promise of ML tools across various aspects of drug formulation science, including both preformulation studies and formulation optimization. Building on these foundational efforts, more advanced data collection and ML techniques are now being integrated, driving innovation and expanding the scope of ML applications in the field. To better understand the trend of breakthroughs in this area, this review examines relevant works published in the past decade, identifying key trends, core applications, and emerging techniques in ML-driven drug delivery. Representative studies are highlighted as examples to illustrate the evolving landscape and practical implementations of these technologies. Furthermore, this review explores forward-looking perspectives, highlighting the convergence of ML with the increasing openness of regulatory bodies, the integration of organoid models, and the advancement of experimental automation.
机器学习(ML)越来越多地应用于药物配方科学,以支持更高效、数据驱动的药物开发策略。越来越多的研究已经证明了ML工具在药物配方科学的各个方面的前景,包括预配方研究和配方优化。在这些基础工作的基础上,现在正在整合更先进的数据收集和机器学习技术,推动创新并扩大机器学习在该领域的应用范围。为了更好地了解这一领域的突破趋势,本文回顾了过去十年发表的相关著作,确定了机器学习驱动给药的关键趋势、核心应用和新兴技术。有代表性的研究被强调为例子,以说明这些技术的发展前景和实际实现。此外,本文还探讨了前瞻性的观点,强调了随着监管机构的日益开放,类器官模型的整合以及实验自动化的进步,机器学习的融合。
{"title":"The growing impact of machine learning on drug formulation science","authors":"Zeqing Bao ,&nbsp;Steven Huynh ,&nbsp;Frantz Le Devedec ,&nbsp;Jo Nguyen ,&nbsp;Christine Allen","doi":"10.1016/j.addr.2025.115761","DOIUrl":"10.1016/j.addr.2025.115761","url":null,"abstract":"<div><div>Machine learning (ML) has increasingly been adopted in drug formulation science to support more efficient, data-driven drug development strategies. A growing number of studies have demonstrated the promise of ML tools across various aspects of drug formulation science, including both preformulation studies and formulation optimization. Building on these foundational efforts, more advanced data collection and ML techniques are now being integrated, driving innovation and expanding the scope of ML applications in the field. To better understand the trend of breakthroughs in this area, this review examines relevant works published in the past decade, identifying key trends, core applications, and emerging techniques in ML-driven drug delivery. Representative studies are highlighted as examples to illustrate the evolving landscape and practical implementations of these technologies. Furthermore, this review explores forward-looking perspectives, highlighting the convergence of ML with the increasing openness of regulatory bodies, the integration of organoid models, and the advancement of experimental automation.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115761"},"PeriodicalIF":17.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145731924","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Emerging nanoparticle-based therapies for pancreatic cancer: Current clinical landscape 新兴的基于纳米颗粒的胰腺癌治疗方法:目前的临床前景
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-12 DOI: 10.1016/j.addr.2025.115760
Ainara Salgado-Pascual , Sara Zalba , Juan José Lasarte , Maria J. Garrido
Pancreatic cancer, particularly ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal subtypes due to late diagnosis, the absence of early biomarkers, and the presence of a complex tumor microenvironment (TME). This TME is characterized by a dense desmoplastic stroma, hypovascularization, immunosuppression, and an acidic extracellular pH. All of these factors hinder the delivery and efficacy of conventional therapies, especially in advanced stages.
Nanoparticles (NPs), including liposomes, polymeric micelles, albumin-bound particles and lipid nanoparticles, have emerged as a promising tool for overcoming TME barriers, and enhance drug delivery in tumor while minimizing systemic toxicity. NPs can exploit mechanisms such as the Enhanced Permeability and Retention (EPR) effect and active targeting. Clinically approved NPs such as Nab-Paclitaxel and liposomal Irinotecan have demonstrated improved pharmacokinetics and therapeutic benefits in PDAC. Furthermore, ongoing clinical trials are exploring novel NP-based strategies such as gene delivery, radiosensitization, immunomodulation and ferroptosis induction. Despite these promising advances, significant challenges remain, including poor tumor penetration, heterogeneity in EPR, and immune recognition of NPs leading to their clearance from bloodstream before reaching the tumor. Innovative solutions such as biomimetic coatings, stimuli-responsive systems and personalized formulations, are being evaluated to enhance NP performance. Standardization of NP characterization and data reporting are essential to accelerating clinical translation. The integration of artificial intelligence and machine learning could further optimize NP design and patient stratification. Overall, nanotechnology represents a crucial frontier of research for developing more effective and personalized pancreatic cancer treatments.
胰腺癌,特别是导管腺癌(PDAC)是最具侵袭性和致死性的亚型之一,由于诊断较晚,缺乏早期生物标志物,以及存在复杂的肿瘤微环境(TME)。这种TME的特征是致密的间质、低血管化、免疫抑制和酸性细胞外ph。所有这些因素都阻碍了常规治疗的传递和疗效,特别是在晚期。纳米颗粒(NPs),包括脂质体、聚合物胶束、白蛋白结合颗粒和脂质纳米颗粒,已经成为克服TME障碍、增强肿瘤药物传递同时最小化全身毒性的有前途的工具。NPs可以利用增强渗透性和保留性(EPR)效应和主动靶向等机制。临床批准的NPs,如nab -紫杉醇和伊立替康脂质体,已经证明改善了PDAC的药代动力学和治疗效果。此外,正在进行的临床试验正在探索新的基于np的策略,如基因传递、放射增敏、免疫调节和铁下垂诱导。尽管取得了这些有希望的进展,但仍然存在重大挑战,包括肿瘤穿透性差、EPR的异质性以及NPs的免疫识别导致其在到达肿瘤之前从血液中清除。人们正在评估诸如仿生涂层、刺激响应系统和个性化配方等创新解决方案,以提高NP性能。NP表征和数据报告的标准化对于加速临床翻译至关重要。人工智能和机器学习的结合可以进一步优化NP设计和患者分层。总的来说,纳米技术代表了研究开发更有效和个性化的胰腺癌治疗的关键前沿。
{"title":"Emerging nanoparticle-based therapies for pancreatic cancer: Current clinical landscape","authors":"Ainara Salgado-Pascual ,&nbsp;Sara Zalba ,&nbsp;Juan José Lasarte ,&nbsp;Maria J. Garrido","doi":"10.1016/j.addr.2025.115760","DOIUrl":"10.1016/j.addr.2025.115760","url":null,"abstract":"<div><div>Pancreatic cancer, particularly ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal subtypes due to late diagnosis, the absence of early biomarkers, and the presence of a complex tumor microenvironment (TME). This TME is characterized by a dense desmoplastic stroma, hypovascularization, immunosuppression, and an acidic extracellular pH. All of these factors hinder the delivery and efficacy of conventional therapies, especially in advanced stages.</div><div>Nanoparticles (NPs), including liposomes, polymeric micelles, albumin-bound particles and lipid nanoparticles, have emerged as a promising tool for overcoming TME barriers, and enhance drug delivery in tumor while minimizing systemic toxicity. NPs can exploit mechanisms such as the Enhanced Permeability and Retention (EPR) effect and active targeting. Clinically approved NPs such as Nab-Paclitaxel and liposomal Irinotecan have demonstrated improved pharmacokinetics and therapeutic benefits in PDAC. Furthermore, ongoing clinical trials are exploring novel NP-based strategies such as gene delivery, radiosensitization, immunomodulation and ferroptosis induction. Despite these promising advances, significant challenges remain, including poor tumor penetration, heterogeneity in EPR, and immune recognition of NPs leading to their clearance from bloodstream before reaching the tumor. Innovative solutions such as biomimetic coatings, stimuli-responsive systems and personalized formulations, are being evaluated to enhance NP performance. Standardization of NP characterization and data reporting are essential to accelerating clinical translation. The integration of artificial intelligence and machine learning could further optimize NP design and patient stratification. Overall, nanotechnology represents a crucial frontier of research for developing more effective and personalized pancreatic cancer treatments.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115760"},"PeriodicalIF":17.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145731034","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploiting colloidal drug aggregation for drug delivery: From promise to prediction using computational tools 利用胶体药物聚集给药:从使用计算工具的承诺到预测
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-11 DOI: 10.1016/j.addr.2025.115758
Kai V. Slaughter , Xiang Olivia Li , Molly S. Shoichet
Colloidal drug aggregates are amorphous nanoparticles formed by the self-assembly of hydrophobic small molecule drugs. They can be leveraged as drug-rich nanoparticle formulations for drug delivery. However, it is difficult to predict which drugs will form colloidal aggregates, which stabilizers will be effective, and what the in vivo fate of the nanoparticles will be. These challenges can be addressed, in part, with computational tools including artificial intelligence such as machine learning. Molecular dynamics simulations have been used to improve our understanding of the intermolecular forces that govern the assembly of colloidal drug aggregates. Several predictive tools exist to identify aggregators, but these are typically used to eliminate aggregators from screening libraries rather than design drug delivery formulations. Colloidal drug aggregates require stabilizers to prevent particle growth and precipitation. Computational analyses have been used to predict which colloidal drug aggregators can be stabilized by a particular small molecule excipient and to identify drug-stabilizer pairs. Successful stabilization has enabled colloidal drug aggregate evaluation for applications such as nanomedicine and sustained release. Additionally, certain colloid-forming drugs can be useful for co-delivery of nucleic acids. In future studies, computational tools can be developed to predict the biological activity of colloidal drug aggregates, building upon other approaches currently used for lipid nanoparticles and other modalities. Ultimately, leveraging computational strategies to improve the design of colloidal drug aggregates can help realize the potential of this high drug-loading delivery platform.
胶体药物聚集体是由疏水小分子药物自组装形成的无定形纳米颗粒。它们可以作为富含药物的纳米颗粒配方用于药物输送。然而,很难预测哪些药物会形成胶体聚集体,哪些稳定剂会有效,以及纳米颗粒在体内的命运如何。这些挑战可以部分地通过包括机器学习等人工智能在内的计算工具来解决。分子动力学模拟已被用于提高我们对控制胶体药物聚集体组装的分子间力的理解。有几种预测工具可以识别聚合器,但这些工具通常用于从筛选库中消除聚合器,而不是设计给药配方。胶体药物聚集体需要稳定剂来防止颗粒生长和沉淀。计算分析已经被用来预测哪些胶体药物聚集剂可以被特定的小分子赋形剂稳定,并识别药物稳定剂对。成功的稳定使胶体药物聚集体的应用评估,如纳米医学和缓释。此外,某些形成胶体的药物可用于核酸的共递送。在未来的研究中,可以开发计算工具来预测胶体药物聚集体的生物活性,以目前用于脂质纳米颗粒和其他模式的其他方法为基础。最终,利用计算策略来改进胶体药物聚集体的设计可以帮助实现这种高载药递送平台的潜力。
{"title":"Exploiting colloidal drug aggregation for drug delivery: From promise to prediction using computational tools","authors":"Kai V. Slaughter ,&nbsp;Xiang Olivia Li ,&nbsp;Molly S. Shoichet","doi":"10.1016/j.addr.2025.115758","DOIUrl":"10.1016/j.addr.2025.115758","url":null,"abstract":"<div><div>Colloidal drug aggregates are amorphous nanoparticles formed by the self-assembly of hydrophobic small molecule drugs. They can be leveraged as drug-rich nanoparticle formulations for drug delivery. However, it is difficult to predict which drugs will form colloidal aggregates, which stabilizers will be effective, and what the <em>in vivo</em> fate of the nanoparticles will be. These challenges can be addressed, in part, with computational tools including artificial intelligence such as machine learning. Molecular dynamics simulations have been used to improve our understanding of the intermolecular forces that govern the assembly of colloidal drug aggregates. Several predictive tools exist to identify aggregators, but these are typically used to eliminate aggregators from screening libraries rather than design drug delivery formulations. Colloidal drug aggregates require stabilizers to prevent particle growth and precipitation. Computational analyses have been used to predict which colloidal drug aggregators can be stabilized by a particular small molecule excipient and to identify drug-stabilizer pairs. Successful stabilization has enabled colloidal drug aggregate evaluation for applications such as nanomedicine and sustained release. Additionally, certain colloid-forming drugs can be useful for co-delivery of nucleic acids. In future studies, computational tools can be developed to predict the biological activity of colloidal drug aggregates, building upon other approaches currently used for lipid nanoparticles and other modalities. Ultimately, leveraging computational strategies to improve the design of colloidal drug aggregates can help realize the potential of this high drug-loading delivery platform.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115758"},"PeriodicalIF":17.6,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145717893","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Overcoming barriers and shaping the future: Challenges and innovations in nucleic acid therapies for Glioblastoma 克服障碍,塑造未来:胶质母细胞瘤核酸治疗的挑战与创新
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-10 DOI: 10.1016/j.addr.2025.115759
Alaa Zam , Nadia Rouatbi , Adam A. Walters , Khuloud T. Al-Jamal
Glioblastoma (GBM) is the most aggressive and treatment-resistant primary brain tumor in adults. Conventional therapies offer limited benefit due to the tumor's heterogeneity, invasive nature, and the presence of the blood–brain barrier, which restricts therapeutic access. Nucleic acid (NA)-based therapies, including small interfering RNA, microRNA, antisense oligonucleotides, splice-switching oligonucleotides, and CRISPR-based systems, have emerged as promising tools to modulate oncogenic pathways and overcome resistance mechanisms at the genetic level. However, effective delivery remains the primary challenge in translating these therapies into clinical success. This review examines the current landscape of NA-based strategies for GBM, with a focus on innovative delivery systems designed to navigate biological barriers and enhance therapeutic precision. We highlight clinical progress made with nanocarrier platforms such as liposomes, lipid nanoparticles, and exosome-based systems, and evaluate their safety, specificity, and delivery efficiency. Additionally, we discuss the most promising preclinical advances, including multifunctional, targeted, and stimuli-responsive carriers, that demonstrate strong potential for clinical translation. Our analysis underscores that the therapeutic efficacy of NA approaches in GBM is inseparable from the sophistication of their delivery platforms. Moving forward, the integration of rationally designed carriers with gene-targeted payloads holds the key to unlocking the full potential of precision medicine in GBM.
胶质母细胞瘤(GBM)是成人中最具侵袭性和治疗耐药性的原发性脑肿瘤。由于肿瘤的异质性、侵袭性和血脑屏障的存在,限制了治疗途径,传统疗法的疗效有限。基于核酸(NA)的治疗,包括小干扰RNA、微RNA、反义寡核苷酸、剪接开关寡核苷酸和基于crispr的系统,已经成为在遗传水平上调节致癌途径和克服耐药机制的有前途的工具。然而,有效的递送仍然是将这些疗法转化为临床成功的主要挑战。本文综述了目前基于na的GBM治疗策略,重点关注旨在克服生物屏障和提高治疗精度的创新给药系统。我们强调了纳米载体平台(如脂质体、脂质纳米颗粒和外泌体系统)的临床进展,并评估了它们的安全性、特异性和递送效率。此外,我们还讨论了最有希望的临床前进展,包括多功能、靶向和刺激反应性载体,这些载体显示出强大的临床转化潜力。我们的分析强调NA入路治疗GBM的疗效与其输送平台的复杂性密不可分。展望未来,整合合理设计的载体与基因靶向有效载荷是释放GBM精准医疗全部潜力的关键。
{"title":"Overcoming barriers and shaping the future: Challenges and innovations in nucleic acid therapies for Glioblastoma","authors":"Alaa Zam ,&nbsp;Nadia Rouatbi ,&nbsp;Adam A. Walters ,&nbsp;Khuloud T. Al-Jamal","doi":"10.1016/j.addr.2025.115759","DOIUrl":"10.1016/j.addr.2025.115759","url":null,"abstract":"<div><div>Glioblastoma (GBM) is the most aggressive and treatment-resistant primary brain tumor in adults. Conventional therapies offer limited benefit due to the tumor's heterogeneity, invasive nature, and the presence of the blood–brain barrier, which restricts therapeutic access. Nucleic acid (NA)-based therapies, including small interfering RNA, microRNA, antisense oligonucleotides, splice-switching oligonucleotides, and CRISPR-based systems, have emerged as promising tools to modulate oncogenic pathways and overcome resistance mechanisms at the genetic level. However, effective delivery remains the primary challenge in translating these therapies into clinical success. This review examines the current landscape of NA-based strategies for GBM, with a focus on innovative delivery systems designed to navigate biological barriers and enhance therapeutic precision. We highlight clinical progress made with nanocarrier platforms such as liposomes, lipid nanoparticles, and exosome-based systems, and evaluate their safety, specificity, and delivery efficiency. Additionally, we discuss the most promising preclinical advances, including multifunctional, targeted, and stimuli-responsive carriers, that demonstrate strong potential for clinical translation. Our analysis underscores that the therapeutic efficacy of NA approaches in GBM is inseparable from the sophistication of their delivery platforms. Moving forward, the integration of rationally designed carriers with gene-targeted payloads holds the key to unlocking the full potential of precision medicine in GBM.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115759"},"PeriodicalIF":17.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145717923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Engineering tumor spatial heterogeneity in vitro 体外工程肿瘤空间异质性
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-10 DOI: 10.1016/j.addr.2025.115757
Changchong Chen , Zixuan Zhao , Dong Hua Seah , Kenny Zhuoran Wu , Senthilkumar Mohanaselvi , Eliza Li Shan Fong
Spatial heterogeneity is a fundamental feature of the tumor microenvironment, characterized by structured variations in cellular composition, phenotypic states, extracellular matrix (ECM) organization, and biochemical and biophysical gradients. These spatial patterns shape tumor evolution, modulate immune infiltration, and underlie resistance to therapy. Advances in spatial transcriptomics and multiplex imaging have revealed dynamic and region-specific niches, such as hypoxic cores, immune-excluded zones, and fibroblast-dense invasive fronts, that correlate with clinical outcomes. However, most in vitro models fail to capture this architectural complexity. Recent engineering technologies, including 3D bioprinting, organoid assembloids, organ-on-a-chip systems, and ECM-mimetic scaffolds, now enable controlled reconstruction of tumor spatial organization and microregional heterogeneity. These technologies allow integration of patient-derived cells, tunable matrix environments, and spatially defined signaling to mimic in vivo pathophysiology. When integrated with spatial transcriptomics and proteomics, these models enable mechanistic exploration of microregional tumor biology, evaluation of therapeutic responses, and investigation of immunotherapy resistance. This review integrates our current understanding of spatial heterogeneity in cancer with enabling engineering strategies to guide future developments in tumor biology and therapeutic innovation.
空间异质性是肿瘤微环境的一个基本特征,其特征是细胞组成、表型状态、细胞外基质(ECM)组织以及生化和生物物理梯度的结构性变化。这些空间模式塑造肿瘤的演变,调节免疫浸润,并对治疗产生抵抗。空间转录组学和多重成像技术的进步揭示了与临床结果相关的动态和区域特异性生态位,如缺氧核心、免疫排斥区和成纤维细胞密集侵入前沿。然而,大多数体外模型无法捕获这种架构复杂性。最近的工程技术,包括3D生物打印、类器官组装体、器官芯片系统和模拟ecm支架,现在可以控制肿瘤空间组织和微区域异质性的重建。这些技术允许整合患者来源的细胞、可调节的基质环境和空间定义的信号来模拟体内病理生理。当与空间转录组学和蛋白质组学相结合时,这些模型可以对微区域肿瘤生物学进行机制探索,评估治疗反应,并研究免疫治疗耐药性。这篇综述将我们目前对癌症空间异质性的理解与工程策略结合起来,以指导肿瘤生物学和治疗创新的未来发展。
{"title":"Engineering tumor spatial heterogeneity in vitro","authors":"Changchong Chen ,&nbsp;Zixuan Zhao ,&nbsp;Dong Hua Seah ,&nbsp;Kenny Zhuoran Wu ,&nbsp;Senthilkumar Mohanaselvi ,&nbsp;Eliza Li Shan Fong","doi":"10.1016/j.addr.2025.115757","DOIUrl":"10.1016/j.addr.2025.115757","url":null,"abstract":"<div><div>Spatial heterogeneity is a fundamental feature of the tumor microenvironment, characterized by structured variations in cellular composition, phenotypic states, extracellular matrix (ECM) organization, and biochemical and biophysical gradients. These spatial patterns shape tumor evolution, modulate immune infiltration, and underlie resistance to therapy. Advances in spatial transcriptomics and multiplex imaging have revealed dynamic and region-specific niches, such as hypoxic cores, immune-excluded zones, and fibroblast-dense invasive fronts, that correlate with clinical outcomes. However, most in vitro models fail to capture this architectural complexity. Recent engineering technologies, including 3D bioprinting, organoid assembloids, organ-on-a-chip systems, and ECM-mimetic scaffolds, now enable controlled reconstruction of tumor spatial organization and microregional heterogeneity. These technologies allow integration of patient-derived cells, tunable matrix environments, and spatially defined signaling to mimic in vivo pathophysiology. When integrated with spatial transcriptomics and proteomics, these models enable mechanistic exploration of microregional tumor biology, evaluation of therapeutic responses, and investigation of immunotherapy resistance. This review integrates our current understanding of spatial heterogeneity in cancer with enabling engineering strategies to guide future developments in tumor biology and therapeutic innovation.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115757"},"PeriodicalIF":17.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145731927","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrating PROTAC-based targeted protein degradation with nanodelivery systems to overcome cancer therapeutic resistance 整合基于protac的靶向蛋白降解与纳米递送系统克服癌症治疗耐药性
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-10 DOI: 10.1016/j.addr.2025.115755
Xinyu Gou , Shi He , Bilan Wang , Lingli Zhang , Yongzhong Cheng , Xiang Gao
Tumor drug resistance is a major challenge in cancer treatment, as traditional chemotherapeutic agents and small molecule inhibitors often become ineffective in targeting tumors due to drug resistance. Proteolysis Targeting Chimeras (PROTAC) technology, as a novel protein degradation method, provides a new insight into overcoming drug resistance in tumors with the assistance of nanodelivery systems. PROTAC is able to degrade rather than merely inhibit tumor-associated proteins, thus avoiding drug resistance caused by gene mutations, protein overexpression and conformational changes, demonstrating significant advantages in overcoming tumor resistance. First, PROTAC eliminates the biological activity of the target protein by directly degrading it, thus overcoming the limitation of traditional inhibitors, which are susceptible to mutations of the structure and activity of the target protein. Second, PROTAC molecules are highly versatile and flexible, and can target proteins that are difficult to target with conventional drugs, including enzymatically inactive proteins, transcription factors and oncogenic protein complexes. In addition, PROTAC technology, with the booster of nanodelivery systems, can effectively improve solubility and bioavailability, enhance targeting and delivery efficiency while improving its stability, and can be combined with other therapeutic methods to further enhance the therapeutic effect. The versatility of PROTAC makes it a highly promising option for overcoming tumor drug resistance, and their effectiveness has been validated in a variety of cancers, including breast cancer, prostate cancer, and leukemia. In this paper, we will review the recent progress of PROTAC technology in overcoming tumor drug resistance and briefly summarize the advantages and challenges of PROTAC technology combined with nanodelivery system, hoping to provide valuable references for researchers in related fields.
肿瘤耐药是肿瘤治疗的主要挑战,传统的化疗药物和小分子抑制剂往往由于耐药而无法靶向肿瘤。蛋白质水解靶向嵌合体(Proteolysis Targeting Chimeras, PROTAC)技术作为一种新的蛋白质降解方法,为利用纳米递送系统克服肿瘤耐药提供了新的思路。PROTAC能够降解而不仅仅是抑制肿瘤相关蛋白,从而避免了基因突变、蛋白过表达和构象改变引起的耐药,在克服肿瘤耐药方面具有显著优势。首先,PROTAC通过直接降解目标蛋白来消除其生物活性,从而克服了传统抑制剂易受目标蛋白结构和活性突变的限制。其次,PROTAC分子具有高度的通用性和灵活性,可以靶向传统药物难以靶向的蛋白质,包括酶失活蛋白、转录因子和致癌蛋白复合物。此外,PROTAC技术在纳米递送系统的助推下,可有效提高溶解度和生物利用度,在提高稳定性的同时增强靶向性和递送效率,并可与其他治疗方法联合使用,进一步提高治疗效果。PROTAC的多功能性使其成为克服肿瘤耐药性的极有希望的选择,其有效性已在多种癌症中得到验证,包括乳腺癌,前列腺癌和白血病。本文将综述PROTAC技术在克服肿瘤耐药方面的最新进展,并简要总结PROTAC技术结合纳米给药系统的优势和挑战,希望为相关领域的研究人员提供有价值的参考。
{"title":"Integrating PROTAC-based targeted protein degradation with nanodelivery systems to overcome cancer therapeutic resistance","authors":"Xinyu Gou ,&nbsp;Shi He ,&nbsp;Bilan Wang ,&nbsp;Lingli Zhang ,&nbsp;Yongzhong Cheng ,&nbsp;Xiang Gao","doi":"10.1016/j.addr.2025.115755","DOIUrl":"10.1016/j.addr.2025.115755","url":null,"abstract":"<div><div>Tumor drug resistance is a major challenge in cancer treatment, as traditional chemotherapeutic agents and small molecule inhibitors often become ineffective in targeting tumors due to drug resistance. Proteolysis Targeting Chimeras (PROTAC) technology, as a novel protein degradation method, provides a new insight into overcoming drug resistance in tumors with the assistance of nanodelivery systems. PROTAC is able to degrade rather than merely inhibit tumor-associated proteins, thus avoiding drug resistance caused by gene mutations, protein overexpression and conformational changes, demonstrating significant advantages in overcoming tumor resistance. First, PROTAC eliminates the biological activity of the target protein by directly degrading it, thus overcoming the limitation of traditional inhibitors, which are susceptible to mutations of the structure and activity of the target protein. Second, PROTAC molecules are highly versatile and flexible, and can target proteins that are difficult to target with conventional drugs, including enzymatically inactive proteins, transcription factors and oncogenic protein complexes. In addition, PROTAC technology, with the booster of nanodelivery systems, can effectively improve solubility and bioavailability, enhance targeting and delivery efficiency while improving its stability, and can be combined with other therapeutic methods to further enhance the therapeutic effect. The versatility of PROTAC makes it a highly promising option for overcoming tumor drug resistance, and their effectiveness has been validated in a variety of cancers, including breast cancer, prostate cancer, and leukemia. In this paper, we will review the recent progress of PROTAC technology in overcoming tumor drug resistance and briefly summarize the advantages and challenges of PROTAC technology combined with nanodelivery system, hoping to provide valuable references for researchers in related fields.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115755"},"PeriodicalIF":17.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145717895","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gas-based therapeutics and delivery platforms in cancer immunotherapy 肿瘤免疫治疗中的气体疗法和输送平台
IF 17.6 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-06 DOI: 10.1016/j.addr.2025.115746
Van-Anh Thi Nguyen , Chieh-Cheng Huang , Yunching Chen
Gas-based therapeutics are emerging as a promising strategy in cancer immunotherapy. Small gaseous signaling molecules such as nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and oxygen (O2) efficiently penetrate tumor tissues and modulate diverse immune pathways. These therapeutic gases can relieve tumor hypoxia, enhance immune cell infiltration, induce immunogenic cancer cell death, and suppress immunosuppressive signaling within the tumor microenvironment (TME). Therefore, they potentiate immune checkpoint blockade and other immunotherapies while overcoming key barriers to immune evasion. Despite this promise, the clinical translation of gas-based therapies faces significant challenges, including short half-lives, systemic toxicity, and lack of spatiotemporal control. To address these limitations, a variety of delivery platforms have been developed—from nanocarriers and injectable hydrogels to inhalable and oral prodrug formulations and stimuli-responsive systems—that enable safe, tumor-targeted, and controlled release of therapeutic gases. Such engineered strategies maximize antitumor efficacy while minimizing off-target effects. This review highlights the immunomodulatory roles of therapeutic gases, examines state-of-the-art delivery technologies, and discusses how these advances lay the foundation for precision gas immunotherapy to unlock the clinical potential of gaseous immunomodulators in cancer treatment.
基于气体的治疗方法正在成为一种有前景的癌症免疫治疗策略。小的气体信号分子如一氧化氮(NO)、一氧化碳(CO)、硫化氢(H2S)和氧(O2)有效地渗透肿瘤组织并调节多种免疫途径。这些治疗气体可以缓解肿瘤缺氧,增强免疫细胞浸润,诱导免疫原性癌细胞死亡,抑制肿瘤微环境(tumor microenvironment, TME)内的免疫抑制信号。因此,它们增强了免疫检查点封锁和其他免疫疗法,同时克服了免疫逃避的关键障碍。尽管前景光明,但气体疗法的临床转化面临着重大挑战,包括半衰期短、全身毒性和缺乏时空控制。为了解决这些限制,已经开发了各种递送平台-从纳米载体和可注射水凝胶到可吸入和口服前药制剂和刺激反应系统-使治疗气体的安全,肿瘤靶向和控制释放。这种工程策略最大限度地提高了抗肿瘤效果,同时最大限度地减少了脱靶效应。这篇综述强调了治疗气体的免疫调节作用,检查了最先进的输送技术,并讨论了这些进步如何为精确气体免疫治疗奠定基础,以释放气体免疫调节剂在癌症治疗中的临床潜力。
{"title":"Gas-based therapeutics and delivery platforms in cancer immunotherapy","authors":"Van-Anh Thi Nguyen ,&nbsp;Chieh-Cheng Huang ,&nbsp;Yunching Chen","doi":"10.1016/j.addr.2025.115746","DOIUrl":"10.1016/j.addr.2025.115746","url":null,"abstract":"<div><div>Gas-based therapeutics are emerging as a promising strategy in cancer immunotherapy. Small gaseous signaling molecules such as nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H<sub>2</sub>S), and oxygen (O<sub>2</sub>) efficiently penetrate tumor tissues and modulate diverse immune pathways. These therapeutic gases can relieve tumor hypoxia, enhance immune cell infiltration, induce immunogenic cancer cell death, and suppress immunosuppressive signaling within the tumor microenvironment (TME). Therefore, they potentiate immune checkpoint blockade and other immunotherapies while overcoming key barriers to immune evasion. Despite this promise, the clinical translation of gas-based therapies faces significant challenges, including short half-lives, systemic toxicity, and lack of spatiotemporal control. To address these limitations, a variety of delivery platforms have been developed—from nanocarriers and injectable hydrogels to inhalable and oral prodrug formulations and stimuli-responsive systems—that enable safe, tumor-targeted, and controlled release of therapeutic gases. Such engineered strategies maximize antitumor efficacy while minimizing off-target effects. This review highlights the immunomodulatory roles of therapeutic gases, examines state-of-the-art delivery technologies, and discusses how these advances lay the foundation for precision gas immunotherapy to unlock the clinical potential of gaseous immunomodulators in cancer treatment.</div></div>","PeriodicalId":7254,"journal":{"name":"Advanced drug delivery reviews","volume":"229 ","pages":"Article 115746"},"PeriodicalIF":17.6,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145689331","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","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
Advanced drug delivery reviews
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