{"title":"Collagen-Anchoring Polymer Dots Enable Rapid and Precise Detection of Microlesions in Inflammatory Bowel Disease through Perivascular Matrix Accumulation.","authors":"Shuting Lu, Yuqiao Li, Zhuang Zhang, Minglei Teng, Jingwen Hou, Xi Tan, Qing Lu, Liqin Xiong","doi":"10.1002/adhm.202501580","DOIUrl":"https://doi.org/10.1002/adhm.202501580","url":null,"abstract":"<p><p>Inflammatory bowel disease (IBD) poses substantial challenges in early diagnosis and the detection of small lesions due to clinical heterogeneity and limitations of existing imaging techniques. In this study, functional polymer dots (Pdots) are developed to rapidly and precisely image microlesions by extravasating from IBD-associated vasculature and anchoring within the perivascular matrix mediated by collagen. These Pdots are engineered with dual-channel fluorescence (visible/NIR windows) and photoacoustic (PA) imaging capabilities, making them suitable for non-invasive IBD diagnosis. Diagnosis using these functional Pdots can be completed within 3 h, which is significantly faster than current nanoprobes. Furthermore, the Pdots allow for continuous visualization of the IBD-affected areas for up to 24 h. They are capable of detecting microlesions smaller than 200 µm, surpassing the resolution achieved in previous studies. Additionally, even when utilizing wide-field stereoscopic fluorescence microscopy, the signal-to-background ratio (SBR) in IBD-affected areas reaches up to 4.75. High-resolution microvascular imaging reveals IBD-associated intestinal vascular remodeling, including mucosal vascular dilation and submucosal pathological angiogenesis at a resolution of 2-3 µm. Notably, IBD induces a marked increase in the proportion of microvessels with diameters less than 20 µm in the cecum, Peyer's patches (PP), and mesenteric lymph nodes (MLN) of mice. This work establishes functional Pdots as promising nanoplatforms for rapid and precise IBD diagnosis by leveraging the pathological features of IBD-affected areas. Moreover, they facilitate real-time and high-resolution visualization of microvasculature, offering significant potential for guiding therapeutic interventions.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e01580"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673430","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yeonjin Kim, Jinseong Kim, Ji Hye Jeong, Inki Kim, Nayeon Shim, Hyeonji Yoo, Eunsung Jun, Kwangmeyung Kim
{"title":"Cancer-Specific and Pro-Apoptotic PEGylated Liposomes Containing SMAC-Mimetic Doxorubicin Prodrug for Safe High-Dose Delivery in Pancreatic Cancer.","authors":"Yeonjin Kim, Jinseong Kim, Ji Hye Jeong, Inki Kim, Nayeon Shim, Hyeonji Yoo, Eunsung Jun, Kwangmeyung Kim","doi":"10.1002/adhm.202502018","DOIUrl":"https://doi.org/10.1002/adhm.202502018","url":null,"abstract":"<p><p>High-dose chemotherapy exhibits potent therapeutic efficacy in pancreatic cancer. However, its application is often limited because of severe toxicity and drug resistance. Herein, a high-dose therapy of pro-apoptotic doxorubicin (DOX) prodrug-encapsulated PEGylated liposomes (Aposomes) is presented for the treatment of pancreatic cancer. The prodrug is synthesized by conjugating DOX with a second mitochondria-derived activator of caspases mimetic peptide (SMAC-P-FRRL; AVPIAQ-FRRL: antagonist of inhibitor of apoptosis proteins), in which the cathepsin B-cleavable -RR- sequence enables selective release. The resulting SMAC-P-FRRL-DOX is efficiently encapsulated into PEGylated liposomes (87.7 ± 0.48 nm), which induced apoptosis specifically in cathepsin B-overexpressing pancreatic cancer cells. In KPC960 tumor-bearing mice, repeated administration of high-dose Aposomes facilitates preferential tumor accumulation via the enhanced permeability and retention (EPR) effect and exerts potent antitumor activity with minimal toxicity in normal tissues. Moreover, the therapeutic efficacy and safety profile of high-dose Aposomes are validated in humanized NOD scid gamma (NSG) mice. Notably, high-dose Aposomes significantly reduce orthotopic pancreatic tumor size in NSG mice without toxicity in the blood, immune system, and normal tissues. This high-dose therapy of Aposomes can greatly improve the therapeutic index of pancreatic cancer chemotherapy without causing severe toxicity and potential drug resistance.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e02018"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yaya Wei, Li Li, Tingting Fu, Ting Gao, Lanlan Ma, Wenbao Zuo, Jianhong Yang
{"title":"Immunological Tolerance Induced by Nanoliposome with Autoantigenie Peptide and Artesunate to Inhibit Complement and Remodel Immune Balance for Multiple Sclerosis Treatment.","authors":"Yaya Wei, Li Li, Tingting Fu, Ting Gao, Lanlan Ma, Wenbao Zuo, Jianhong Yang","doi":"10.1002/adhm.202403774","DOIUrl":"https://doi.org/10.1002/adhm.202403774","url":null,"abstract":"<p><p>Multiple sclerosis (MS) is a demyelinating autoimmune disease (AD) accompanied by immune disorders and complement over activation. Although re-instatement of immune balance does alleviate MS symptoms, exploration of concomitant complement inhibition for neuron protection has not been evaluated. Herein, we developed myelin oligodendrocyte glycoprotein 35-55 (MOG) peptide and artemisinin (ART) co-loaded liposomes (MOG-ART-Lip) to simultaneously restore immune balance and inhibit complement activation. This nanoplatform enhanced solubility of both components while enabling CNS delivery. Liposomes loaded with MOG would induce tolerogenic DCs (tol-DCs) that express low-levels of costimulatory molecules, capable of antigenic peptide presentation and induction of regulatory T cells, while ART shifted microglia from pro-inflammatory (M1) to anti-inflammatory (M2) phenotypes. Importantly, ART suppressed complement-mediated demyelination via the C3/C3a receptor (C3aR) pathway. In vivo studies showed MOG-ART-Lip significantly reduced neuroinflammation, attenuated demyelination, and promoted neural repair, leading to functional recovery. Overall, results of this study suggest that a combination of an auto-antigenic peptide and an immune-modulator provides a promising modality for the treatment of MS by re-establishing antigen-specific immune tolerance. As such, the results of the study provide valuable insight into a new approach for development of combinatorial complement therapies for the treatment of MS patients.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2403774"},"PeriodicalIF":10.0,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144657916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Boming Peng, Min Huang, Jianquan Zhang, Yang Xiang
{"title":"Advances in Biliary Disease Organoid Research: From Model Construction to Clinical Applications.","authors":"Boming Peng, Min Huang, Jianquan Zhang, Yang Xiang","doi":"10.1002/adhm.202501776","DOIUrl":"https://doi.org/10.1002/adhm.202501776","url":null,"abstract":"<p><p>The biliary system is vital to hepatobiliary function, yet diseases like primary sclerosing cholangitis, biliary atresia, cystic fibrosis, and cholangiocarcinoma remain poorly understood due to the limitations of traditional two-dimensional (2D) cell cultures and animal models, which fail to replicate complex human biliary physiology. Biliary organoids, an innovative three-dimensional (3D) in vitro model, have emerged to bridge this gap, closely mimicking tissue structure and function. This review systematically summarizes the construction methods of biliary organoids-matrix-independent methods and matrix-dependent methods, as well as tissue engineering-based strategies, such as bioprinting and microfluidics-and cell sources, including primary tissues, pluripotent stem cells, and tumor-derived cells. It also explores the potential roles of key signaling pathways that drive biliary development and disease in guiding cell differentiation, proliferation, and tissue organization during biliary organoid construction. It explores recent applications in disease modeling and clinical translation, leveraging gene editing, chemical induction, inflammatory stimulation, and co-culture systems. Despite their potential, challenges persist in model stability, long-term culture, and immune microenvironment simulation. Future advances, integrating multi-omics, dynamic culture systems, and emerging bioengineering technologies, promise to enhance physiological relevance. Biliary organoids are poised to transform fundamental research, drug screening, and personalized medicine, accelerating clinical breakthroughs in hepatobiliary disease management.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2501776"},"PeriodicalIF":10.0,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144657915","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rui Ding, Hongyu Yang, Jiaxin Wang, Yuejuan Liu, Sen Mu, Dongkai Wang, Ji Li
{"title":"Advances in Stimuli-Responsive Release Strategies for Sonosensitizers in Synergistic Sonodynamic Immunotherapy against Tumors.","authors":"Rui Ding, Hongyu Yang, Jiaxin Wang, Yuejuan Liu, Sen Mu, Dongkai Wang, Ji Li","doi":"10.1002/adhm.202502183","DOIUrl":"https://doi.org/10.1002/adhm.202502183","url":null,"abstract":"<p><p>Despite intensive efforts to develop diagnostic and therapeutic tools, the successful treatment of cancer is still hampered by uncontrolled tumor proliferation and metastasis. Sonodynamic therapy (SDT) is a cutting-edge, noninvasive treatment modality that activates sonosensitizers via low-intensity ultrasound (US) to generate reactive oxygen species (ROS), offering deep tissue penetration, low phototoxicity, and minimal side effects. Beyond direct cytotoxicity, it can trigger immunogenic cell death (ICD), thereby enhancing systemic antitumor immune responses. However, its efficacy is constrained by the immunosuppressive tumor microenvironment (ITME). To address these limitations, growing research efforts have focused on dual therapeutic strategies that combine SDT with tumor immunotherapy. These strategies are designed to enhance tumor-specific accumulation of sonosensitizers through stimuli-responsive release mechanisms. SDT activates immune pathways and induces ICD, thus remodeling the tumor microenvironment (TME) and converting immunologically \"cold\" tumors into \"hot\" tumors. This transformation effectively addresses the low response rates and immune-related adverse events associated with immunotherapy, while enhancing immune recognition and tumor clearance. This review summarizes recent advances in the development of stimuli-responsive release strategies for sonosensitizers in sonodynamic immunotherapy, discusses the challenges hindering clinical translation, and underscores the potential of this dual therapeutic strategy in advancing cancer treatment.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2502183"},"PeriodicalIF":10.0,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144641301","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Recent Advances in Dual-Function Janus Membranes for Guided Periodontal and Bone Regeneration.","authors":"Ying Li, Yeying Lin, Tianhua Xiao, Wen Liu, Chengyun Ning, Guoxin Tan, Lei Zhou","doi":"10.1002/adhm.202502888","DOIUrl":"https://doi.org/10.1002/adhm.202502888","url":null,"abstract":"<p><p>Guided Tissue Regeneration (GTR) and Guided Bone Regeneration (GBR) are essential surgical techniques in periodontal therapy, employing barrier membranes to prevent soft tissue infiltration and create a conducive environment for bone regeneration. However, the regenerative performance of conventional barrier membranes remains limited due to poor interface management and insufficient biological functionality. Recent developments have introduced the concept of Janus membranes-structures with asymmetric, dual-function surfaces-offering promising solutions to these challenges. While various reviews have addressed barrier membranes for periodontal and bone regeneration, comprehensive reviews specifically focusing on multifunctional Janus membranes are still limited. This review highlights recent advances in Janus membrane design for GTR and GBR applications. It first outlines key structural configurations, followed by an in-depth analysis of fabrication techniques and functional strategies, including osteogenesis promotion, antibacterial activity, and immunomodulation. By summarizing current progress and challenges, this review offers valuable insights into next-generation biomaterial development for periodontal regeneration. Looking forward, Janus membranes represent a compelling avenue for enhancing clinical outcomes in GTR and GBR procedures.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2502888"},"PeriodicalIF":10.0,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144648036","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Van-Nghia Nguyen, Thuy Giang Nguyen Cao, Hyunsun Jeong, Quan Truong Hoang, Binh T. T. Pham, Jieun Bang, Chang Woo Koh, Ji Hee Kang, Jeong Hyun Lee, Xiaofeng Wu, Won Jong Rhee, Young Tag Ko, K. M. K. Swamy, Sungnam Park, JaeHong Park, Min Suk Shim, Juyoung Yoon
{"title":"Tumor-Targeted Exosome-Based Heavy Atom-Free Nanosensitizers With Long-Lived Excited States for Safe and Effective Sono-Photodynamic Therapy of Solid Tumors (Adv. Healthcare Mater. 18/2025)","authors":"Van-Nghia Nguyen, Thuy Giang Nguyen Cao, Hyunsun Jeong, Quan Truong Hoang, Binh T. T. Pham, Jieun Bang, Chang Woo Koh, Ji Hee Kang, Jeong Hyun Lee, Xiaofeng Wu, Won Jong Rhee, Young Tag Ko, K. M. K. Swamy, Sungnam Park, JaeHong Park, Min Suk Shim, Juyoung Yoon","doi":"10.1002/adhm.202570106","DOIUrl":"https://doi.org/10.1002/adhm.202570106","url":null,"abstract":"<p><b>Sono-Photodynamic Cancer Therapy</b></p><p>In article 2500927, JaeHong Park, Min Suk Shim, Juyoung Yoon, and co-workers introduce tumor-targeted exosomes encapsulating a heavy atom-free nanosensitizer (IR820-TPE) for fluorescence imaging-guided sono-photodynamic cancer therapy. The IR820-TPE-loaded exosomes efficiently generate ROS via both type I and II pathways and eradicate tumors under near-infrared light and ultrasound irradiation.\u0000\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":"14 18","pages":""},"PeriodicalIF":10.0,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/adhm.202570106","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144634975","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hongjuan Weng, Monize Caiado Decarli, Lei He, Wen Chen, Sabine van Rijt, Katrien V Bernaerts, Lorenzo Moroni
{"title":"Mechanical Reinforced and Self-healing Hydrogels: Bioprinted Biomimetic Methacrylated Collagen Peptide-Xanthan Gum Constructs for Ligament Regeneration.","authors":"Hongjuan Weng, Monize Caiado Decarli, Lei He, Wen Chen, Sabine van Rijt, Katrien V Bernaerts, Lorenzo Moroni","doi":"10.1002/adhm.202502341","DOIUrl":"https://doi.org/10.1002/adhm.202502341","url":null,"abstract":"<p><p>Collagen peptide (COP) is water soluble, bioactive, and tends to be a promising alternative to collagen for tissue regeneration. However, its low viscosity and lack of readily polymerizable groups hinder its bioprinting and limit its wide applications in tissue engineering. In this study, methacrylated collagen peptide-xanthan gum (COPMA-XG) bioinks with interpenetrating networks are developed for bioprinting stable constructs, followed by stem cell differentiation. First, self-healing COPMA hydrogels are developed with rapid UV-curing and tunable mechanical properties. To increase the printability and the mechanical properties of COPMA, XG is mixed to create a set of COPMA-XG bioinks. COPMA-XG hydrogels show self-healing properties, optimal printability, and stable morphology in the medium. The bioprinted human bone marrow mesenchymal stem cells (hMSCs) laden COPMA-XG constructs are biocompatible and bioactive, with increased production of extracellular matrix, collagen type I, and scleraxis over 28 days. Overall, bioprinted COPMA-XG constructs are versatile matrices to support hMSCs proliferation and differentiation with potential for ligament tissue engineering.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2502341"},"PeriodicalIF":10.0,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144641302","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}