Wang Xiang, Suisui He, Zhi Zheng, Ying Liu, Xinping Liu, Tao Kuang, Zongtao Zhou, Jun Wang, Cui-Yun Yu, Hua Wei
{"title":"A Self-Adaptive Rhodium(I) Complex-Based Nanoplatform with Type II Immunogenic Cell Death for Near-Infrared Phosphorescence Imaging and Cancer Immunotherapy.","authors":"Wang Xiang, Suisui He, Zhi Zheng, Ying Liu, Xinping Liu, Tao Kuang, Zongtao Zhou, Jun Wang, Cui-Yun Yu, Hua Wei","doi":"10.1002/adhm.202501871","DOIUrl":"https://doi.org/10.1002/adhm.202501871","url":null,"abstract":"<p><p>Despite immunogenic cell death (ICD) has garnered significant attention in cancer therapy, achieving precise in vivo immunity activation and simultaneous visualization of immunotherapy processes remain significant challenges due to the difficulties in facile integration of multifunctionalities in a single nanomedicine. For this purpose, herein a self-adaptive rhodium(I) complex-based nanoplatform driven by metallophilic interactions is reported not only for near-infrared (NIR) imaging-guided cancer immunotherapy, but also as the first example of a rhodium(I)-based ICD inducer. Specifically, this nanoplatform enables high tumor enrichment by utilizing homologous targeting capability camouflaged by cancer cell membranes and facilitates enhanced in vivo NIR phosphorescence imaging. The subsequent uptake of this nanoplatform by tumor cells via endocytosis releases the antitumor rhodium(I) complex monomer, which can target directly the endoplasmic reticulum and induce a more effective type II ICD for enhanced dendritic cell maturation and cytotoxic T lymphocyte infiltration, and ultimately lead to long-acting antitumor immunity. Notably, the self-adaptive functional switch strongly supports the NIR phosphorescence imaging and cancer immunotherapy of this platform, which displays a remarkable inhibitory effect with a tumor inhibition rate of 91.2%. This study develops a facile yet robust approach toward an \"all-in-one\" metal-based ICD agent with visualization properties for monitoring immunotherapy.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e01871"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673426","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":"Boosting Sonodynamic/Gas/Chemo Therapy through Triple Inhibiting Multidrug Resistance Using Responsive Biodegradable Sulfide-Vacancy-Rich Nanosheets.","authors":"Dongmiao Cao, Wei Xia, Kaiyang Wang, Aihong Chen, Ruixin Luo, Lile Dong, Jie Lu, Yicheng Zhu, Xuebo Yin, Yu Luo, Xijian Liu","doi":"10.1002/adhm.202502378","DOIUrl":"https://doi.org/10.1002/adhm.202502378","url":null,"abstract":"<p><p>Non-targeted chemotherapy remains the primary therapeutic approach for treating triple-negative breast cancer (TNBC), but it frequently results in multidrug resistance and severe side effects. In this study, a responsive biodegradable zinc-doped MoS<sub>2-x</sub> nanosheet is developed with rich sulfide vacancies (ZMS) to enhance sono-chemotherapy in TNBC by simultaneously inhibiting multidrug resistance through triple-pathway modulation (reactive oxygen species, H<sub>2</sub>S, and Zn<sup>2+</sup>). The introduction of sulfide-vacancies via Zn doping significantly inhibits electron-hole recombination, and eventually boosts the generation of reactive oxygen species under ultrasound (US) activation to enhance sonodynamic therapy (SDT). In addition, the ZMS/DOX can degrade in an acidic tumor microenvironment (TME) to release DOX (doxorubicin hydrochloride), Zn<sup>2+</sup>, and H<sub>2</sub>S. Zn<sup>2+</sup> inhibits intracellular ATP production by disrupting glycolysis in cancer cells, while H<sub>2</sub>S synergistically reduces intracellular ATP levels by impairing the mitochondrial electron transport chain. Furthermore, the reduction in ATP levels suppresses the expression of P-glycoprotein, thereby overcoming drug resistance. Additionally, ZMS exhibits catalase-like activity to convert H<sub>2</sub>O<sub>2</sub> into O<sub>2</sub> in TME, relieving the tumor's hypoxia as well as enhancing the therapeutic efficacy of SDT and chemotherapy. The proposed biodegradable therapeutic platform holds great promise for strengthening sono-chemotherapy in TNBC treatment and overcoming the limitations associated with traditional chemotherapy.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e02378"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673428","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":"Photonic Energy Back Transfer for Enhanced Upconversion/NIR-II Luminescence with 3D-Printed Manufacturing for Bone Imaging.","authors":"Yanxing Wang, Ziyue Ju, Ji Zhang, Yuji Li, Fanbo Meng, Weiwei He, Ruichan Lv","doi":"10.1002/adhm.202503246","DOIUrl":"https://doi.org/10.1002/adhm.202503246","url":null,"abstract":"<p><p>In this study, cubic-phase YOF is first identified as a suitable luminescent host for rare earth nanoparticle (RENP) through molecular dynamics simulations. By optimizing the core-shell structure, doping elements, and their ratios in the nanoparticles, the enhancement effect of energy back transfer (EBT) from Nd<sup>3+</sup> to Er<sup>3+</sup> on Nd<sup>3+</sup> near-infrared-II (NIR-II) luminescence and Er<sup>3+</sup> upconversion luminescence (UCL) is discovered. By separating the emission elements of NIR-II luminescence and UCL, energy competition is avoided, enabling the simultaneous enhancement of NIR-II luminescence and two-photon UCL. Based on this optimized NIR-II emission, high-quality in vivo vascular and bone imaging in mice are achieved. Finally, the potential of this optimized RENP as a novel bone material is explored using 3D printing technology. The proposed SiO<sub>2</sub>-RENP multilayer structure effectively prevents laser-induced ejection of pure RENP while enhancing biocompatibility and mechanical properties. Through in vivo implantation experiments, this multilayer material demonstrates excellent long-term stability and biocompatibility in the NIR-II window, indicating its great potential as a novel bone material.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e03246"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673431","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}
Thomas Meslier, Justin Christopher D'Antin, Gemma Julio, Anna Roig
{"title":"A Comprehensive Review of Clinical Studies on Bacterial Cellulose: From the Earliest Uses to Contemporary Innovations.","authors":"Thomas Meslier, Justin Christopher D'Antin, Gemma Julio, Anna Roig","doi":"10.1002/adhm.202502189","DOIUrl":"https://doi.org/10.1002/adhm.202502189","url":null,"abstract":"<p><p>Bacterial cellulose (BC), a biopolymer produced by Gram-negative bacteria such as those from the Komagataeibacter genus, has emerged as a promising material in biomedical applications due to its exceptional purity, biocompatibility, mechanical strength, versatility in sterilization, biodegradability, and sustainable production. This comprehensive review traces the evolution of BC-based medical products from their initial development in the 1980s to present-day innovations, highlighting the progression of clinical evidence supporting their use. Early clinical studies primarily focused on BC as a temporary skin substitute for burns and ulcers. Over time, the scope of BC applications expanded to include dural defect repair, tympanic membrane reconstruction, and fetal surgery for spina bifida. The quality of clinical evidence has improved, with recent years seeing an increase in randomized controlled trials and larger patient cohorts. Several BC-based products are already commercially available, supported by regulatory approvals and a growing body of clinical data. This review seeks to serve as a valuable resource by providing an exhaustive recapitulation of published clinical studies on BC-based medical devices encompassing 3,857 subjects and offering insights into their efficacy, safety, and regulatory considerations. The review outlines future research directions to validate BC's potential across a wide range of in vivo biomedical applications.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e02189"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673425","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":"Accurate Neurotransmitter Release by Ultrasound-Responsive Nanomaterials for Alleviating L-DOPA-Induced Dyskinesia in Parkinson's Disease.","authors":"Shengwei Jia, Xiaoxia Li, Fang Wang, Hao Zhang, Qun-Dong Shen","doi":"10.1002/adhm.202501765","DOIUrl":"https://doi.org/10.1002/adhm.202501765","url":null,"abstract":"<p><p>Functional nanomaterials show great promise in next-generation targeted non-genetic neuromodulation and on-demand neurotransmitter release with superior spatiotemporal accuracy. L-DOPA-induced dyskinesia (LID) is a common complication during the treatment process of Parkinson's disease. Dysfunction in the dynamics of dopamine release plays a pivotal role in the disease. A strategy for LID treatment by using ultrasound-responsive functional nanomaterials for controlled release of nitric oxide is proposed, which can activate the nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) signaling pathway. This alleviates the abnormal synaptic plasticity caused by excessive and pulsatile dopamine, and diminishes hypersensitivity of the D<sub>1</sub> dopamine receptors, thereby reducing the behavioral disorders associated with LID. These findings offer a novel perspective for therapy of neurodegenerative diseases.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e01765"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673427","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}
Mohammad Nasrullah, Remant Kc, Amarnath Praphakar Rajendran, Saba Abbasi Dezfouli, Cezary Kucharski, Xiaoyan Jiang, Spencer B Gibson, Joseph Brandwein, Olaf Heidenreich, Hasan Uludağ
{"title":"Suppressing t(4;11) Acute Leukemia by Lipopolymer Nanoparticle Delivery of siRNA Targeting KMT2A::AFF1 with Enhanced Extrahepatic Delivery.","authors":"Mohammad Nasrullah, Remant Kc, Amarnath Praphakar Rajendran, Saba Abbasi Dezfouli, Cezary Kucharski, Xiaoyan Jiang, Spencer B Gibson, Joseph Brandwein, Olaf Heidenreich, Hasan Uludağ","doi":"10.1002/adhm.202502019","DOIUrl":"https://doi.org/10.1002/adhm.202502019","url":null,"abstract":"<p><p>Effective siRNA delivery in acute lymphoblastic leukemia (ALL) is limited by preferential hepatic accumulation. To address this, a lipopolymer (PEI-C) is developed by conjugating lipid to polyethylenimine and formulated lipopolymer nanoparticles (LPNPs) via complexation with siRNA. The siRNA delivery efficiency of LPNPs is evaluated in vitro in t(4;11)-positive ALL cells (RS4;11 and SEM) as well as \"normal\" peripheral blood mononuclear cells (PBMCs) from human donors and bone marrow stromal cells (BMSCs) from mice. Molecular effects are assessed by quantifying target mRNA silencing and downstream apoptosis. In vivo biodistribution and therapeutic efficacy are examined in mouse models. LPNPs demonstrated significantly higher siRNA uptake than commercial reagents in PBMCs and BMSCs, reaching siRNA uptakes of 87.2% and 93.0% in RS4;11 and SEM cells, respectively. Molecular analyses revealed effective silencing of KMT2A::AFF1 mRNA (≈80% in RS4;11), accompanied by BCL2 downregulation and increased apoptosis. In vivo, LPNPs showed efficient siRNA biodistribution to leukemia-associated organs (spleen and bone marrow) and significantly reduced leukemia burden in a systemic RS4;11 xenograft mouse model and improved survival. These findings suggest that PEI-C-formulated LPNPs present a promising avenue for therapeutic siRNA delivery in ALL, effectively targeting leukemia-associated organs, and warrant further exploration in clinical studies.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e02019"},"PeriodicalIF":10.0,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673432","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":"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}