Aaron C. Davis , Siting Zhang , Adalyn Meeks , Diya Sakhrani , Luis Carlos Sanjuan Acosta , D. Ethan Kelley , Emma Caldwell , Luis Solorio , Craig J. Goergen , David J. Cappelleri
{"title":"Novel design of 3D printed tumbling microrobots for in vivo targeted drug delivery","authors":"Aaron C. Davis , Siting Zhang , Adalyn Meeks , Diya Sakhrani , Luis Carlos Sanjuan Acosta , D. Ethan Kelley , Emma Caldwell , Luis Solorio , Craig J. Goergen , David J. Cappelleri","doi":"10.1016/j.jconrel.2025.114271","DOIUrl":"10.1016/j.jconrel.2025.114271","url":null,"abstract":"","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114271"},"PeriodicalIF":11.5,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145216058","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}
Fei Han, Linzhou Yin, Lihang Qu, Shuwen Han, Mengchi Sun, Yang Xi
{"title":"Microorganism delivery of cancer vaccines","authors":"Fei Han, Linzhou Yin, Lihang Qu, Shuwen Han, Mengchi Sun, Yang Xi","doi":"10.1016/j.jconrel.2025.114288","DOIUrl":"https://doi.org/10.1016/j.jconrel.2025.114288","url":null,"abstract":"Cancer immunotherapeutic vaccines offer a promising approach which can activate the immune system to target and kill cancer cells. Despite great progresses, challenges still exist in clinical practice, including immunosuppressive tumor microenvironment, tumor heterogeneity as well as low delivery efficiency. Microbe-based cancer vaccines, leveraging the abilities of bacteria and viruses to target hypoxic tumor regions/infect cancer cells as well as enhance the immunogenicity of tumor microenvironment, have attached prominent attention for the potential to revolutionize vaccine designing. Particularly, advances in synthetic biology enable precise expression of tumor antigens or immune checkpoint inhibitors in microbial cancer vaccines, furthermore facilitating personalized immunotherapeutic potential. This review describes the interactions between microorganisms and the host immune system, and systematically introduces the tumor-targeting mechanisms of microorganisms. Specifically, we provide a comprehensive describe about advantages and applications of bacteria, as a type of microbial vaccines, for antitumor treatment. We also delve into multiple kinds of viral vaccines and virus-like particles for cancer therapy. Recent examples of combining microbial vaccines with other therapeutic modalities designed to combat tumors are highlighted. Last but not least, we address the underlying challenges of microbial cancer vaccines. Meanwhile, this review prospects that microbial cancer vaccines represents a transformative immunotherapy strategy with tumor-targeting and immune-activating potential.","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"92 1","pages":""},"PeriodicalIF":10.8,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203239","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}
{"title":"Nanoparticle-driven reactive oxygen species therapy: A new frontier in osteosarcoma treatment","authors":"Lei He, Pamela Habibovic, Sabine van Rijt","doi":"10.1016/j.jconrel.2025.114289","DOIUrl":"10.1016/j.jconrel.2025.114289","url":null,"abstract":"<div><div>Reactive oxygen species (ROS) play a critical role in osteosarcoma (OS) therapy by inducing programmed cell death when their levels exceed the cellular threshold. Due to their inherently elevated ROS levels, OS cells are more susceptible to ROS upregulation than normal bone cells, offering a therapeutic window for selective cancer cell elimination. However, the intrinsic antioxidant defense mechanisms in OS cells mitigate the effects of conventional ROS-generating agents, limiting their clinical efficacy. To overcome this challenge, nanoparticle-based strategies have been developed to enhance ROS production and improve therapeutic outcomes. Despite significant progress, a comprehensive framework for designing and optimizing ROS-generating nanoplatforms for OS treatment remains lacking. This review systematically classifies ROS-upregulating nanoparticles based on their underlying mechanisms of action and discusses their therapeutic potential. Additionally, key challenges and future directions are highlighted to guide the development of next-generation nanomaterials for OS therapy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114289"},"PeriodicalIF":11.5,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203240","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}
Shengwei Lu , Yunqiu Miao , Dandan Wang , Dan Xu , Ruichi Liu , Xinrui Liu , Yang Zhang , Xinxin Zhang , Huanlong Qin
{"title":"Engineered tumor-symbiotic bacterial membrane nanovesicles enable precise immuno-chemotherapy of colorectal cancer","authors":"Shengwei Lu , Yunqiu Miao , Dandan Wang , Dan Xu , Ruichi Liu , Xinrui Liu , Yang Zhang , Xinxin Zhang , Huanlong Qin","doi":"10.1016/j.jconrel.2025.114291","DOIUrl":"10.1016/j.jconrel.2025.114291","url":null,"abstract":"<div><div>Gut microorganisms show promising therapeutic effects and drug delivery potential for colorectal cancer (CRC) treatment, but are limited by their insufficient targeting ability and side effects. <em>Fusobacterium nucleatum</em> (Fn) is a key symbiotic bacterium in CRC, which can preferentially accumulate in tumor tissues and invade tumor cells, while its tumorigenicity restricts the application in drug delivery. Herein, we engineered Fn with anchored PD-L1 antibody (αPD-L1), and then isolated the Fn membranes to construct bacterial membrane nanovesicles (ab-FMNVs) for precise delivery of chemotherapeutic drugs. The ab-FMNVs exploited Fn's inherent tumor colonization capabilities to achieve tumor-targeted delivery through the specific membrane protein FadA-mediated pathway, and modulated the PD-L1 immune checkpoint pathway for tumor immunotherapy. Simultaneously, ab-FMNVs were internalized into CT26 cells to release the chemotherapeutic agent doxorubicin, synergistically inhibiting tumor cell proliferation and metastasis. In a CRC-bearing mouse model, doxorubicin-loaded ab-FMNVs increased tumor accumulation and demonstrated superior antitumor efficacy against both primary and recurrent CRC progression without inducing any side effects. This innovative approach holds promise for precision cancer therapies by harnessing the symbiotic relationship between bacteria and CRC.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114291"},"PeriodicalIF":11.5,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145209775","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}
Hui Wang , Liang Qi , Yue-Fei Fang , Yang Liu , Zheng Ye , Bi-Te Chen , Yi Wang , Hu-Lin Jiang , Ling Li
{"title":"Fibrotic collagen-targeted delivery system blocks pancreatic intercellular crosstalk to alleviate pancreatic fibrosis","authors":"Hui Wang , Liang Qi , Yue-Fei Fang , Yang Liu , Zheng Ye , Bi-Te Chen , Yi Wang , Hu-Lin Jiang , Ling Li","doi":"10.1016/j.jconrel.2025.114278","DOIUrl":"10.1016/j.jconrel.2025.114278","url":null,"abstract":"<div><div>Pancreatic fibrosis (PF) is a significant disruption of homeostasis in the pancreas, primarily characterized by excessive extracellular matrix (ECM) deposition due to pancreatic acinar cell (PAC) injury, pancreatic stellate cell (PSC) activation, and persistent inflammatory response. As a hallmark feature of both chronic pancreatitis and pancreatic cancer, progressive fibrosis exacerbates disease severity and poses significant clinical challenges. Notably, the self-amplifying crosstalk between activated PSCs and injured PACs perpetuates fibrogenesis and undermines therapeutic efficacy. However, currently, no effective strategy is available to modulate intercellular and alleviate pancreatic fibrosis. Herein, we developed dual drug-loaded lipid nanoparticles (JM-CCs) functionalized with a collagen-binding peptide (CBP) and collagenase I on their surface. This design facilitates targeted drug delivery by enabling penetration through the dense ECM barrier to the core of fibrotic lesions. The released melatonin alleviates oxidative stress in PACs, thereby reducing their fibrogenic stimulation of PSCs. Concurrently, the encapsulated JTE013 suppresses PSC activation by modulating autophagy, leading to decreased ECM production and mitigation of PAC injury. In a caerulein-induced PF mouse model, JM-CCs effectively reduce ECM deposition and repair pancreatic exocrine function. This study provides a novel strategy for regaining pancreatic tissue homeostasis and offers a promising approach for therapies of fibrosis and pancreatic disease.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114278"},"PeriodicalIF":11.5,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203242","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}
Hangyu Zhou , Xueping Luo , Chuyu Huang , Yixin Liu , Qiuyuan Li , Guangmiao Chen , Rongrong Zheng , Shiying Li , Linping Zhao
{"title":"A dual PROTAC nanocarrier amplifies DNA damage and STING activation for cancer immunotherapy","authors":"Hangyu Zhou , Xueping Luo , Chuyu Huang , Yixin Liu , Qiuyuan Li , Guangmiao Chen , Rongrong Zheng , Shiying Li , Linping Zhao","doi":"10.1016/j.jconrel.2025.114290","DOIUrl":"10.1016/j.jconrel.2025.114290","url":null,"abstract":"<div><div>The cytoplasmic accumulation of damaged DNA to activate the STING pathway has emerged as a promising strategy to enhance tumor immunogenicity and improve the efficacy of immune checkpoint blockade (ICB) therapy. Herein, a stimuli-responsive and dual PROTAC-embedded immunoactivator (denoted as Sd@Lip) is developed to potentiate ICB through amplified DNA damage and robust STING activation. Sd@Lip comprises an acid-sensitive liposomal nanocarrier co-encapsulating two targeted degraders of dBET1 (a BRD4 degrader) and SK-575 (a PARP1 degrader), which enhances drug solubility, stability, and enable precise stoichiometric co-delivery. Mechanistically, Sd@Lip enhances DNA damage by simultaneously disrupting both nonhomologous end joining (NHEJ) and homologous recombination (HR) repair pathways, leading to cytoplasmic DNA accumulation that activates STING signaling, induces proinflammatory cytokine release and enhances infiltration of immune effector cells. This immunogenic cascade promotes the recruitment of natural killer (NK) cells and cytotoxic T lymphocytes into the tumor microenvironment, thereby significantly augmenting the therapeutic efficacy of ICB against both primary and metastatic breast tumors. Collectively, this study highlights a synergistic PARP1 and BRD4 degradation strategy to induce immunostimulatory DNA damage, offering a compelling approach to improve outcomes in breast cancer immunotherapy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114290"},"PeriodicalIF":11.5,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203243","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}
Xuyang Ning , Hong Lu , Huiying Zeng , Ziqiang Zhou , Ping Hu
{"title":"Multifunctional self-healing hydrogels for chronic wound repair: Design, mechanisms, and applications","authors":"Xuyang Ning , Hong Lu , Huiying Zeng , Ziqiang Zhou , Ping Hu","doi":"10.1016/j.jconrel.2025.114282","DOIUrl":"10.1016/j.jconrel.2025.114282","url":null,"abstract":"<div><div>Chronic wounds arise from a multifactorial interplay involving persistent hyperglycemia, recurrent infection, tissue ischemia, and sustained inflammation, which collectively disrupt cellular metabolic homeostasis, impair angiogenesis and extracellular matrix remodeling, ultimately leading to impaired healing progression. Multifunctional self-healing hydrogels have become promising biomaterials to solve these problems due to their excellent biocompatibility, self-healing ability and multiple functional properties. These hydrogels can autonomously restore their structure and function after injury, thus maintaining the therapeutic effect of the wound site. In addition, they exhibit multifunctional properties, including antimicrobial activity, adhesion, hemostasis, anti-inflammatory and antioxidant effects, drug delivery, promotion of angiogenesis and nerve repair, electrical conductivity, and monitoring capabilities, resulting in significantly improved wound healing outcomes. In this paper, the healing process and current status of chronic wounds are first discussed, and then the latest progress in the preparation mechanism, functional properties and mechanical properties of multifunctional self-healing hydrogels is studied. The application of these hydrogels in chronic wound healing is critically analyzed, with a focus on their self-healing mechanisms and functional properties,and it is worth emphasizing the first comprehensive analysis of the hydrogels' function in neurovascular regeneration. In addition, the design related to clinical translation is also discussed. Future research directions are proposed, including the development of smart hydrogels and the exploration of new biomaterials to provide more innovative and effective solutions for chronic wound management. Overall, multifunctional self-healing hydrogels represent a transformative approach to chronic wound healing with great potential for clinical applications.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114282"},"PeriodicalIF":11.5,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203208","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}
{"title":"Lipid and polymeric nanocarriers for siRNA delivery to the brain","authors":"Sushil Koirala, Kun Cheng","doi":"10.1016/j.jconrel.2025.114286","DOIUrl":"10.1016/j.jconrel.2025.114286","url":null,"abstract":"<div><div>Brain diseases refer to any pathological conditions that impair the normal functions of the brain. These mainly include neurodegenerative disorders, brain injuries, and malignant tumors. Researchers have explored various therapeutic approaches to treat these conditions, with gene modulation gaining significant attention in recent years due to its potential to address the root cause of disease. One promising approach is the use of siRNAs as therapeutic agents for brain diseases. Most preclinical studies on siRNA delivery to the brain have employed invasive intracerebral methods to target specific brain regions, posing significant challenges for clinical translation. The challenges associated with the intracerebral route highlight the need for safer, more practical, and patient-compliant alternatives, such as systemic delivery. However, systemic siRNA delivery faces obstacles due to its inherent instability in circulation and the restrictive nature of the blood-brain barrier (BBB). Nanocarriers have emerged as a promising strategy to overcome these challenges. These nanocarriers can be made from various natural, synthetic, or biological materials, with polymers and lipids being the most commonly used due to their biocompatibility, ease of surface modification for targeting, controlled drug release, and improved stability. In this review, we discuss lipid- and polymer-based nanotechnology strategies aimed at overcoming the challenges of siRNA delivery and enhancing its therapeutic potential for treating brain diseases.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114286"},"PeriodicalIF":11.5,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203241","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}
Rong Yang , Fangqing Deng , Zibo Gao , Xu Li , Shuaiqi Huangfu , Qing Tian , Haoyu Wang , Huifang Liu , Xuejing Wang , Yao Chen , Yingchun Yang , Genyang Cheng , Lianbing Zhang
{"title":"Melanoma MHC-I-membrane-encapsulated Cu@ferrihydrite induces ferroptosis/cuproptosis and systematic immunity against tumor","authors":"Rong Yang , Fangqing Deng , Zibo Gao , Xu Li , Shuaiqi Huangfu , Qing Tian , Haoyu Wang , Huifang Liu , Xuejing Wang , Yao Chen , Yingchun Yang , Genyang Cheng , Lianbing Zhang","doi":"10.1016/j.jconrel.2025.114281","DOIUrl":"10.1016/j.jconrel.2025.114281","url":null,"abstract":"<div><div>Major histocompatibility complex I (MHC-I) has significant potential for augmenting cancer immunogenicity and immune recognition. Here, we report an innovative therapeutic strategy that synergistically integrates blue light-upregulated MHC-I expression with blue light-induced ferroptosis and cuproptosis. Blue light promoted MHC-I expression in mouse melanoma cells by modulating the NF-κB-SUSD6 signaling axis. Subsequently, an MHC-I-enriched melanoma cytomembrane was used to encapsulate the photoresponsive Cu@ferrihydrite (Cu@Fh) nanoparticles, forming M-Cu@Fh. MHC-I facilitated dendritic cells (DCs) maturation and CD8<sup>+</sup>/CD4<sup>+</sup> T cells activation. M-Cu@Fh also triggered oxidative stress and concurrent ferroptosis/cuproptosis through the controllable release of Fe/Cu ions under blue-light irradiation. <em>In vivo</em> experiments demonstrated that the combination of blue light and M-Cu@Fh converted immune “cold” tumors into “hot” tumors, suppressed <em>in situ</em> melanoma growth through oxidative damages and enhanced immunogenicity. Furthermore, systemic activation of DCs and CD8<sup>+</sup>/CD4<sup>+</sup> T cells in lymphoid organs (lymph nodes and spleen) and lungs conferred prophylactic efficacy against abscopal metastasis. Our study elucidates the photoregulatory mechanism of MHC-I in melanoma cells and presents a transformative combinatorial strategy that synergizes blue light-driven photoimmunotherapy (PIT) with blue light-activated photodynamic therapy (PDT) for melanoma management and metastasis prevention.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"388 ","pages":"Article 114281"},"PeriodicalIF":11.5,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145194998","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}