Li Li, Wang Wang, Rong Li, Jiayue Guo, Xulin Hu, Yu Pan, Taoyuan Zhang, Sensen Chi, Zili Gu, Gaohui Zhu, Qi Liu, Shuai Tan
{"title":"An adeno-associated virus gene therapy strategy for anti-obesity treatment by nanocarrier-based delivery systems.","authors":"Li Li, Wang Wang, Rong Li, Jiayue Guo, Xulin Hu, Yu Pan, Taoyuan Zhang, Sensen Chi, Zili Gu, Gaohui Zhu, Qi Liu, Shuai Tan","doi":"10.1186/s12951-025-03595-5","DOIUrl":"10.1186/s12951-025-03595-5","url":null,"abstract":"<p><p>Gut microbiota is increasingly recognized for its profound influence on host metabolism. However, the mechanisms underlying the distinct metabolic phenotype observed in germ-free (GF) mice are not fully understood. Here, the serum levels of metabolic hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) were shown with significant elevation in GF than the conventionally-raised (CONV-R) mice. Single-cell transcriptome analysis revealed that Secretogranin II (Scg2), with a known function in secretion control, was exclusively expressed in enteroendocrine cells (EECs). Scg2 transcript levels were significantly up-regulated in GF mice, positively correlated with enhanced GLP-1 and peptide PYY secretion. To examine the functional significance of Scg2 in hormone regulation, cross-linked nanoparticles capable of long-term adhesion to intestinal epithelium were designed, with AAV adsorbed within the cross-linked structure. This innovative design enhances its stability and retention in vivo, providing a robust platform for continuous and efficient gene delivery. We evidenced that over-expression of Scg2 via AAV-loaded nanocarriers in the colons of mice on high-fat diet or with the ob/ob genotype not only enhanced the secretion of GLP-1 and PYY but also mitigated weight gain in these mice by reducing their appetite. A multi-omics analysis reveals that Scg2 overexpression in the colon decreased hypothalamic inflammation and activated tryptophan metabolic pathways. Collectively, our findings suggest a potential therapeutic approach for treating metabolic disorders by enhancing Scg2 expression in colonic EECs.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"528"},"PeriodicalIF":10.6,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12278491/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144674985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Wei Sun, Wei Xu, Miaomiao Xiao, Xinge Zhang, Jing Chen, Jinzhe Zhang, Liqun Yang, Quan Na
{"title":"Enhancing exosome stability and delivery with natural polymers to prevent intrauterine adhesions and promote endometrial regeneration: a review.","authors":"Wei Sun, Wei Xu, Miaomiao Xiao, Xinge Zhang, Jing Chen, Jinzhe Zhang, Liqun Yang, Quan Na","doi":"10.1186/s12951-025-03603-8","DOIUrl":"10.1186/s12951-025-03603-8","url":null,"abstract":"<p><p>Intrauterine adhesions (IUAs) are defined as the formation of fibrotic scar tissue within the uterine cavity. They can lead to a range of complications, including hypomenorrhea, amenorrhea, infertility, and recurrent pregnancy loss. Conventional methods of adhesiolysis frequently demonstrate high recurrence rates and suboptimal therapeutic outcomes. Moreover, conventional postoperative IUA prevention strategies (e.g., physical barriers and hormonal therapy) are ineffective and have adverse effects, emphasizing the need for new therapies. Natural polymers are ideal biomaterials for barrier membranes and drug delivery in uterine repair due to their safety, biodegradability, and compatibility. Exosomes, tiny cell-released vesicles containing healing factors (like miRNAs and proteins), help reduce inflammation and scarring while promoting tissue regeneration. Emerging evidence suggests that combining natural polymers with exosomes could provide a potential therapeutic approach for IUAs and endometrial regeneration. This review presents a comprehensive overview of the biological characteristics of natural polymers and exosomes, elucidating their mechanisms of action in endometrial repair. It also provides an in-depth analysis of the current research landscape pertaining to the combined delivery of natural polymers and exosomes in the prevention of IUAs and the promotion of endometrial regeneration.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"529"},"PeriodicalIF":10.6,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12278673/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144674987","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Prunus mume derived extracellular vesicle-like particles alleviate experimental colitis via disrupting NEK7-NLRP3 interaction and inflammasome activation.","authors":"Qi Lv, Hongqiong Yang, Ying Xie, Xinjie Huang, Zhiqi Yan, Yingshan Lv, Yifan Cui, Lihong Hu, Hongzhi Qiao","doi":"10.1186/s12951-025-03567-9","DOIUrl":"10.1186/s12951-025-03567-9","url":null,"abstract":"<p><p>Edible plant derived extracellular vesicle-like particles (EVLPs) have garnered attention as potential therapeutic agents for chronic inflammatory diseases. Prunus mume (PM) is a functional fruit known for its gastrointestinal benefits, yet the detail material basis and potential mechanism remain unclear. Here, we reported that oral administration of prunus mume derived EVLPs (PM-EVLPs) substantially mitigated experimental colitis in mice. The in vivo bio-distribution analysis revealed that PM-EVLPs specifically targeted inflamed colon of colitis mice. Further in vitro studies demonstrated that PM-EVLPs were predominantly internalized by macrophages. The combined treatment with clodronate liposomes confirmed that macrophage was the target cell for PM-EVLPs-mediated anti-colitis activity. Mechanistically, PM-EVLPs selectively inhibited caspase-1 auto-cleavage and IL-1β secretion caused by NLRP3 inflammasome activation, while exerting minimal impact on AIM2, NLRP1 or NLRC4 inflammasome activation. Excluding the effects on mitochondrial ROS generation, K<sup>+</sup> efflux or Ca<sup>2+</sup> influx, PM-EVLPs disrupted the NEK7-NLRP3 interaction, thereby preventing NLRP3 inflammasome assembly. Notably, the inhibitory activity was attributed to RNAs rather than lipids or proteins within PM-EVLPs. Deep RNA sequencing, coupled with the application of miRNA mimics/inhibitors identified miR159 as the material basis for PM-EVLPs' inhibition of NLRP3 inflammasome activation and anti-colitis efficacy. Collectively, these findings suggest that PM-EVLPs represent a promising nanomedicine with potential as a novel therapeutic strategy for colitis and deserves further investigation and development.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"532"},"PeriodicalIF":10.6,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12278495/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144674998","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jian Guan, Fanhe Meng, Chuanzhuo Wang, Binglun Zhang, Jun Chen, Jingnan Han
{"title":"Recent advances in engineered exosome-based therapies for ocular vascular disease.","authors":"Jian Guan, Fanhe Meng, Chuanzhuo Wang, Binglun Zhang, Jun Chen, Jingnan Han","doi":"10.1186/s12951-025-03589-3","DOIUrl":"10.1186/s12951-025-03589-3","url":null,"abstract":"<p><p>Ocular neovascular diseases (ONDs), including corneal neovascularization (CoNV), age-related macular degeneration (AMD) and diabetic retinopathy (DR), are among the leading causes of visual impairment worldwide. Current therapeutic strategies predominantly involve intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) agents, which, despite their efficacy, present significant limitations such as drug resistance, frequent intravitreal injections, and insufficient addressing of underlying pathological mechanisms. This review critically examines recent advancements in the application of exosomes as innovative drug delivery platforms for treating ocular neovascular diseases. Exosomes, naturally occurring extracellular vesicles, exhibit superior biocompatibility, low immunogenicity, and intrinsic targeting capabilities, making them ideal carriers for bioactive molecules including proteins, RNAs, and small drugs. We explore the mechanistic roles of exosomes in modulating pathological angiogenesis, inflammation, and tissue repair within the ocular environment. Additionally, this review addresses the current challenges hindering the clinical translation of these exosomes, including large-scale production, regulatory hurdles, and safety concerns. Future perspectives highlight the potential integration of nanoparticles and exosomes with existing therapies, the development of multifunctional and personalized treatment strategies, and the necessity for standardized protocols to facilitate their transition from bench to bedside. By overcoming these challenges, exosomes hold the promise of revolutionizing the therapeutic landscape for ocular neovascular diseases, ultimately enhancing patient outcomes and quality of life.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"526"},"PeriodicalIF":10.6,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12276678/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144667738","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fei Cao, Yuandong Xu, Yupeng Guan, Kexin Zhang, Haowei Qiu, Zhen Xu, Yunru He, Ze Xiu Xiao, Gao-Feng Zha, Jun Pang
{"title":"Enhancing the potency of 5T4 mRNA vaccine by CD70 mRNA-LNPs through ADCC and T cell boosting in prostate cancer therapy.","authors":"Fei Cao, Yuandong Xu, Yupeng Guan, Kexin Zhang, Haowei Qiu, Zhen Xu, Yunru He, Ze Xiu Xiao, Gao-Feng Zha, Jun Pang","doi":"10.1186/s12951-025-03607-4","DOIUrl":"10.1186/s12951-025-03607-4","url":null,"abstract":"<p><strong>Background: </strong>Prostate cancer remains a significant health challenge, as conventional treatments often fail to provide long-term benefits in the advanced stages. This study explored the potential of mRNA vaccines as novel therapeutic strategies, specifically focusing on the combination of 5T4 and CD70 mRNA delivered via lipid nanoparticles (LNPs). 5T4, which is highly expressed in various tumors but minimally expressed in normal tissues, is an ideal target for immunotherapy. CD70 was selected for its capacity to enhance T cell activation and augment the overall immune response.</p><p><strong>Results: </strong>Our findings demonstrated that the combination of 5T4 and CD70 mRNA-LNPs significantly enhanced both humoral and cellular immunity, resulting in improved tumor suppression and prolonged survival in a prostate cancer mouse model. Mechanistically, these LNPs increased the immune responses, including elevated 5T4-specific antibodies, enhanced CD8<sup> +</sup> T cell activity, and more active natural killer (NK) cells.</p><p><strong>Conclusions: </strong>These results suggest that this combined mRNA vaccine strategy may offer more effective and durable treatments for prostate cancer and that CD70 mRNA, as an immune activator, has broader potential for cancer immunotherapy.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"523"},"PeriodicalIF":10.6,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12273320/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144659445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Rapid diagnostic imaging and targeted immunotoxin delivery in aggressive prostate cancer using CEACAM5-specific nanobodies.","authors":"Zhaoming Xiao, Jingbo Ma, Jinpeng Cen, Tao Xie, Liuhai Zheng, Guangwei Shi, Zhifen Li, Yang Li, Chengming Qu, Yuanqiao He, Chong Wang, Jun Xiao, Haibo Jiang, Zhijie Li, Jigang Wang, Shan-Chao Zhao","doi":"10.1186/s12951-025-03600-x","DOIUrl":"10.1186/s12951-025-03600-x","url":null,"abstract":"<p><p>Aggressive variant prostate cancer (AVPC) originates from metastatic prostate cancer (mPCa) following androgen receptor-targeted therapies, leading to diverse pathological subtypes, notably castration-resistant prostate cancer (CRPC). Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), is consistently expressed across AVPC phenotypes, including neuroendocrine prostate carcinoma (NEPC) and double-negative prostate carcinoma (DNPC), which are significant subtypes of CRPC, making it a promising therapeutic target. In this study, A high-affinity nanobody, B12, specific to CEACAM5, was discovered through phage library screening. B12 exhibited robust binding capabilities, enhanced tumor accumulation, and effective tissue penetration, facilitating rapid in vivo imaging of AVPC. The conjugation of B12 with PE38 to create the immunotoxin B12-PE38 showed significant anti-tumor activity in AVPC xenograft models, including one that mimics bone metastasis. When B12-PE38 was combined with docetaxel, it elicited enhanced tumor inhibitory effects, effectively inhibiting tumor progression. This study underscores CEACAM5 as a target for precise imaging and targeted therapy in AVPC, introducing novel diagnostic and therapeutic strategies for a disease that currently faces a dearth of effective treatment options due to the scarcity of well-defined targets.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"525"},"PeriodicalIF":10.6,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12275256/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144667737","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Delivery of an ERK inhibitor using bioactive lipid nanoparticles reduces angiogenesis and prevents oral squamous cell carcinoma development.","authors":"Zixian Huang, Junyue Fang, Li Lin, Nvlue Cai, Siyu Chen, Gui He, Yuan Cao, Guo Wu, Yuepeng Wang, Wende Li, Zhiquan Huang, Phei Er Saw","doi":"10.1186/s12951-025-03577-7","DOIUrl":"10.1186/s12951-025-03577-7","url":null,"abstract":"<p><p>Extracellular regulated protein kinases (ERK) signaling is aberrantly activated in oral squamous cell carcinoma (OSCC), and targeting ERK signaling with ERK1/2 inhibitors is a potential strategy for OSCC treatment. However, methods for the more efficient delivery of ERK inhibitors to improve drug utilization remains a pressing challenge. Notably, the application of antiangiogenic therapies to treat OSCC has received increasing attention, yet single therapies often have very limited efficacy. The combination of antiangiogenic therapies with strategies targeting ERK1/2 signaling for the treatment of OSCC is very promising. In this study, we utilized bioactive phospholipids (dipalmitoyl phosphatidic acid, DPPA) with antiangiogenic functions to encapsulate a small-molecule ERK inhibitor (called NP-AE) and constructed an intrinsically biotherapeutically active nanomedicine delivery platform with dual therapeutic efficacy. NP-AE effectively inhibited the proliferation and promoted the apoptosis of OSCC cell lines. A tongue orthotopic xenograft model and a patient-derived xenograft (PDX) model of OSCC were subsequently used to investigate its therapeutic effect. The nanoplatform was able to accumulate in tumor tissues and was internalized by tumor cells after intravenous administration. NP-AE effectively inhibited ERK1/2 phosphorylation and Angptl2 and VEGFA expression in OSCC in vitro and in vivo and significantly inhibited the growth of xenografts. Taken together, these findings suggest that targeting ERK1/2 signaling in combination with antiangiogenic therapy by NP-AE may be a promising strategy for the treatment of OSCC.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"524"},"PeriodicalIF":10.6,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12273460/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144667736","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Zhuoting Lu, Tong Wang, Xiangchi Xu, Qinglian Hu, Lei Wang, Yuanxiang Jin
{"title":"Carrier-free nano-integrated strategy of phototherapy and chemotherapy for synergetic AKT targeted therapy to inhibit cancer metastasis and recurrence.","authors":"Zhuoting Lu, Tong Wang, Xiangchi Xu, Qinglian Hu, Lei Wang, Yuanxiang Jin","doi":"10.1186/s12951-025-03539-z","DOIUrl":"10.1186/s12951-025-03539-z","url":null,"abstract":"<p><p>A key challenge in clinical anticancer treatments is metastasis and recurrence, to address this challenge, it is crucial to develop combination treatment strategies that target different molecular pathways, considering the high complexity of tumors, as well as to create efficient drug delivery systems that enhance therapeutic efficacy and minimize systemic toxicity. Herein, two clinical small molecular drugs indocyanine green (ICG) and paclitaxel (PTX) were self-assembled into relatively stable, carrier-free nanoparticles (IP NPs) through a simple one-step nanoprecipitation method. The spherical IP NPs demonstrate excellent aqueous stability, tumor-targeted accumulation, and potent apoptosis induction. Combined with AKT inhibitor MK-2206, they effectively modulate survival pathways, suppress metastasis, and prevent tumor recurrence in rechallenge models. This synergistic approach achieves equivalent therapeutic efficacy at reduced doses, enhancing treatment safety. Encouraged by its effective therapy and sensitivity-enhancing properties, this study might show significant potential for malignant tumor synergistic therapy.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"520"},"PeriodicalIF":10.6,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12269299/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144649704","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bo Liao, Yu Tian, Mengtong Guan, Wang Han, Weiwei Yi, Kaiting Li, Xiaoliang Yang, Yajuan Niu, Bin Zhang, Peiyu Teng, Dingqun Bai, Liang Kuang, Ying Zhu, Xiaoyu Han
{"title":"Exosomes derived from platelet-rich plasma alleviate synovial inflammation by enhancing synovial lymphatic function.","authors":"Bo Liao, Yu Tian, Mengtong Guan, Wang Han, Weiwei Yi, Kaiting Li, Xiaoliang Yang, Yajuan Niu, Bin Zhang, Peiyu Teng, Dingqun Bai, Liang Kuang, Ying Zhu, Xiaoyu Han","doi":"10.1186/s12951-025-03591-9","DOIUrl":"10.1186/s12951-025-03591-9","url":null,"abstract":"<p><strong>Background: </strong>Synovial inflammation is a pivotal factor in the pathogenesis of osteoarthritis (OA). Platelet-rich plasma-derived Exosomes (PRP-Exos), known for their low immunogenicity, have demonstrated efficacy in modulating chondrocyte function. However, the specific effects and mechanisms of PRP-Exos in synovial inflammation remain unclear. This study aimed to investigate the therapeutic effects and mechanisms of PRP-Exos in synovial inflammation induced by destabilization of the medial meniscus (DMM) in mice.</p><p><strong>Results: </strong>PRP-Exos were extracted via ultracentrifugation. In vivo experiments revealed that PRP-Exos alleviated pain behaviors and synovial inflammation in DMM mice. Furthermore, it was discovered that PRP-Exos enhanced the synovial lymphatic function in DMM mice and promoted lymphangiogenesis. Meanwhile, the therapeutic effect of PRP-Exos on synovial inflammation was attenuated after inhibition of lymphatic function. In vitro studies demonstrated that PRP-Exos enhanced the proliferation, migration, and tube formation ability of lymphatic endothelial cells (LECs), via regulating the PI3K/Akt signaling pathway.</p><p><strong>Conclusions: </strong>This research is the first to reveal that PRP-Exos alleviate pain behaviors and synovial inflammation in DMM mice through activation of the PI3K/Akt signaling pathway in LECs, thereby enhancing synovial lymphatic function and promoting the clearance of inflammatory cells and associated cytokines. These findings offer a novel theoretical foundation for the treatment of synovial inflammation and other inflammation-associated disorders.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"522"},"PeriodicalIF":10.6,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12269200/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144649707","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Juan Yue, Qian Mei, Panyong Wang, Peng Miao, Wen-Fei Dong, Li Li
{"title":"Correction to: Light-triggered multifunctional nanoplatform for efficient cancer photo-immunotherapy.","authors":"Juan Yue, Qian Mei, Panyong Wang, Peng Miao, Wen-Fei Dong, Li Li","doi":"10.1186/s12951-025-03584-8","DOIUrl":"10.1186/s12951-025-03584-8","url":null,"abstract":"","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"517"},"PeriodicalIF":10.6,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12265144/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144642824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}