首页 > 最新文献

Journal of Controlled Release最新文献

英文 中文
Controlled delivery of HIF-1α via extracellular vesicles with collagen-binding activity for enhanced wound healing
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-08 DOI: 10.1016/j.jconrel.2025.02.010
Sungmi Jeon , Seongeon Cho , Seongkyeong Yoo , Yeji Lee , Jiyoung Goo , Yu Jin Jeong , Gi-Hoon Nam , Hyun-Tae Shin , Jong-Wan Park , Cherlhyun Jeong , Sang Wha Kim , Iljin Kim , In-San Kim
Chronic wounds are often characterized by prolonged inflammation, impaired angiogenesis, and dysregulated hypoxic response, partly caused by the insufficient activation of hypoxia-inducible factor-1 alpha (HIF-1α). This study investigated the potential of engineered extracellular vesicles (EVs) to deliver a stable, constitutively active form of HIF-1α (scHIF-1α) to promote wound healing. A collagen-binding domain (CBD) was integrated into EVs to enhance their retention at wound sites, and collagen sponges were employed as scaffolds to ensure sustained, localized release of scHIF-1α EVs. In vitro studies have demonstrated that scHIF-1α EVs significantly improved cell proliferation, migration, and angiogenesis in dermal fibroblasts, endothelial cells, and keratinocytes—key cells involved in the wound healing process. In vivo, scHIF-1α EVs accelerated wound closure, enhanced tissue regeneration, and promoted angiogenesis in various wound healing models, including excisional wounds, surgical skin flaps, and diabetic wounds. The integration of CBD further enhanced EV retention, amplifying therapeutic outcomes. These results propose that scHIF-1α delivery via EVs, particularly when combined with collagen-based sustained-release systems, offers a promising and patient-friendly therapeutic strategy for treating chronic wounds.
{"title":"Controlled delivery of HIF-1α via extracellular vesicles with collagen-binding activity for enhanced wound healing","authors":"Sungmi Jeon ,&nbsp;Seongeon Cho ,&nbsp;Seongkyeong Yoo ,&nbsp;Yeji Lee ,&nbsp;Jiyoung Goo ,&nbsp;Yu Jin Jeong ,&nbsp;Gi-Hoon Nam ,&nbsp;Hyun-Tae Shin ,&nbsp;Jong-Wan Park ,&nbsp;Cherlhyun Jeong ,&nbsp;Sang Wha Kim ,&nbsp;Iljin Kim ,&nbsp;In-San Kim","doi":"10.1016/j.jconrel.2025.02.010","DOIUrl":"10.1016/j.jconrel.2025.02.010","url":null,"abstract":"<div><div>Chronic wounds are often characterized by prolonged inflammation, impaired angiogenesis, and dysregulated hypoxic response, partly caused by the insufficient activation of hypoxia-inducible factor-1 alpha (HIF-1α). This study investigated the potential of engineered extracellular vesicles (EVs) to deliver a stable, constitutively active form of HIF-1α (scHIF-1α) to promote wound healing. A collagen-binding domain (CBD) was integrated into EVs to enhance their retention at wound sites, and collagen sponges were employed as scaffolds to ensure sustained, localized release of scHIF-1α EVs. In vitro studies have demonstrated that scHIF-1α EVs significantly improved cell proliferation, migration, and angiogenesis in dermal fibroblasts, endothelial cells, and keratinocytes—key cells involved in the wound healing process. In vivo, scHIF-1α EVs accelerated wound closure, enhanced tissue regeneration, and promoted angiogenesis in various wound healing models, including excisional wounds, surgical skin flaps, and diabetic wounds. The integration of CBD further enhanced EV retention, amplifying therapeutic outcomes. These results propose that scHIF-1α delivery via EVs, particularly when combined with collagen-based sustained-release systems, offers a promising and patient-friendly therapeutic strategy for treating chronic wounds.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"380 ","pages":"Pages 330-347"},"PeriodicalIF":10.5,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143350855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A rapamycin-loading platelet membrane hybrid liposome with anti-inflammation effect and long-lasting repair capability for acute kidney injury.
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-08 DOI: 10.1016/j.jconrel.2025.02.012
Miaomiao Zhang, Yang Li, Tongyang Xu, Botao Liu, Yue Liu, Xu Cheng, Junjie Pan, Jianxin Wang

Acute kidney injury (AKI) represents a rapid decline in kidney function, often associated with significant morbidity and mortality. Inefficient management of acute-phase inflammation and inadequate repair of established damage exacerbate AKI and facilitate its progression to chronic kidney disease (CKD). Platelet membrane (PM) has emerged as a promising targeting ligand in various studies. PM proteins can also facilitate the recruitment and differentiation of CD34+ cells (hematopoietic stem cells and endothelial progenitor cells) through both direct and indirect mechanisms, including enhancing adhesion of CD34+ cells to damaged tissues and elevating stromal cell-derived factor-1 (SDF-1) levels in ischemia-reperfusion injury (IRI) kidneys. In parallel, extensive research has demonstrated that rapamycin shows high potential as an anti-inflammatory therapy for AKI. Herein, we design a PM hybrid rapamycin liposome (Rapa@PM-Lipo), which not only improves the delivery efficiency of rapamycin, but also leverages the potential of PM to achieve long-lasting repair. Rapa@PM-Lipo significantly reduced Acute renal Tubular Necrosis (ATN) score in IRI kidneys following intravenous administration, both as a single and multiple doses. This study exploits the therapeutic potential of PM and explores its novel applications for facilitating tissue repair, presenting a promising strategy for the treatment of AKI and mitigating its progression to CKD.

{"title":"A rapamycin-loading platelet membrane hybrid liposome with anti-inflammation effect and long-lasting repair capability for acute kidney injury.","authors":"Miaomiao Zhang, Yang Li, Tongyang Xu, Botao Liu, Yue Liu, Xu Cheng, Junjie Pan, Jianxin Wang","doi":"10.1016/j.jconrel.2025.02.012","DOIUrl":"https://doi.org/10.1016/j.jconrel.2025.02.012","url":null,"abstract":"<p><p>Acute kidney injury (AKI) represents a rapid decline in kidney function, often associated with significant morbidity and mortality. Inefficient management of acute-phase inflammation and inadequate repair of established damage exacerbate AKI and facilitate its progression to chronic kidney disease (CKD). Platelet membrane (PM) has emerged as a promising targeting ligand in various studies. PM proteins can also facilitate the recruitment and differentiation of CD34<sup>+</sup> cells (hematopoietic stem cells and endothelial progenitor cells) through both direct and indirect mechanisms, including enhancing adhesion of CD34<sup>+</sup> cells to damaged tissues and elevating stromal cell-derived factor-1 (SDF-1) levels in ischemia-reperfusion injury (IRI) kidneys. In parallel, extensive research has demonstrated that rapamycin shows high potential as an anti-inflammatory therapy for AKI. Herein, we design a PM hybrid rapamycin liposome (Rapa@PM-Lipo), which not only improves the delivery efficiency of rapamycin, but also leverages the potential of PM to achieve long-lasting repair. Rapa@PM-Lipo significantly reduced Acute renal Tubular Necrosis (ATN) score in IRI kidneys following intravenous administration, both as a single and multiple doses. This study exploits the therapeutic potential of PM and explores its novel applications for facilitating tissue repair, presenting a promising strategy for the treatment of AKI and mitigating its progression to CKD.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":""},"PeriodicalIF":10.5,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143391014","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
Biomimetic calcium-chelation nanoparticles reprogram tumor metabolism to enhance antitumor immunity 仿生钙螯合纳米颗粒重编程肿瘤代谢,增强抗肿瘤免疫。
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-08 DOI: 10.1016/j.jconrel.2025.01.046
Zheng Wang , Ming Wu , Yingmeng Jiang , Junjie Zhou , Sai Chen , Qizhi Wang , Honghao Sun , Yueyang Deng , Zhanwei Zhou , Minjie Sun
Metabolic reprogramming within the tumor microenvironment poses a significant obstacle to the therapeutic efficacy of antitumor immunity. Here, inspired by the diverse programme of cholesterol metabolism between tumor and immune cells, a biocompatible carboxy-modified cyclodextrin carrier equipped with a biomimetic surface was developed to encapsulate FX11 and Avasimibe (RM-CDC@FX11&Ava) for synergistic antitumor metabolic therapy and immunotherapy. Through the manipulation of calcium levels using poly-carboxylic compounds to initiate cholesterol biosynthesis, RM-CDC@FX11&Ava dynamically regulates glycolysis and blocks cholesterol esterification to navigate metabolic reprogramming. The resultant cholesterol augmentation triggered by RM-CDC@FX11&Ava could not only specifically induce 34.3 % tumor cell apoptosis but also promote 57.8 % dendritic cell maturation for antigen presentation and improve the effector function of T cells. Furthermore, the tumor immunosuppressive microenvironment was also reprogrammed by impairing Treg cells through the blockade of lactic acid. As a result, RM-CDC@FX11&Ava showed superior antitumor efficacy in mastadenoma and melanoma models.
肿瘤微环境中的代谢重编程是影响抗肿瘤免疫治疗效果的重要障碍。在这里,受肿瘤和免疫细胞之间胆固醇代谢的不同程序的启发,开发了一种具有仿生表面的生物相容性羧基修饰环糊精载体,用于封装FX11和Avasimibe (RM-CDC@FX11&Ava),用于协同抗肿瘤代谢治疗和免疫治疗。通过使用多羧基化合物操纵钙水平来启动胆固醇生物合成,RM-CDC@FX11&Ava动态调节糖酵解和阻断胆固醇酯化,从而引导代谢重编程。RM-CDC@FX11和ava引起的胆固醇升高不仅能特异性诱导34.3% %的肿瘤细胞凋亡,还能促进57.8% %的树突状细胞成熟以供抗原呈递,提高T细胞的效应功能。此外,肿瘤免疫抑制微环境也被重新编程,通过乳酸阻断损害Treg细胞。结果,RM-CDC@FX11和ava在乳腺腺瘤和黑色素瘤模型中显示出优越的抗肿瘤疗效。
{"title":"Biomimetic calcium-chelation nanoparticles reprogram tumor metabolism to enhance antitumor immunity","authors":"Zheng Wang ,&nbsp;Ming Wu ,&nbsp;Yingmeng Jiang ,&nbsp;Junjie Zhou ,&nbsp;Sai Chen ,&nbsp;Qizhi Wang ,&nbsp;Honghao Sun ,&nbsp;Yueyang Deng ,&nbsp;Zhanwei Zhou ,&nbsp;Minjie Sun","doi":"10.1016/j.jconrel.2025.01.046","DOIUrl":"10.1016/j.jconrel.2025.01.046","url":null,"abstract":"<div><div>Metabolic reprogramming within the tumor microenvironment poses a significant obstacle to the therapeutic efficacy of antitumor immunity. Here, inspired by the diverse programme of cholesterol metabolism between tumor and immune cells, a biocompatible carboxy-modified cyclodextrin carrier equipped with a biomimetic surface was developed to encapsulate FX11 and Avasimibe (RM-CDC@FX11&amp;Ava) for synergistic antitumor metabolic therapy and immunotherapy. Through the manipulation of calcium levels using poly-carboxylic compounds to initiate cholesterol biosynthesis, RM-CDC@FX11&amp;Ava dynamically regulates glycolysis and blocks cholesterol esterification to navigate metabolic reprogramming. The resultant cholesterol augmentation triggered by RM-CDC@FX11&amp;Ava could not only specifically induce 34.3 % tumor cell apoptosis but also promote 57.8 % dendritic cell maturation for antigen presentation and improve the effector function of T cells. Furthermore, the tumor immunosuppressive microenvironment was also reprogrammed by impairing Treg cells through the blockade of lactic acid. As a result, RM-CDC@FX11&amp;Ava showed superior antitumor efficacy in mastadenoma and melanoma models.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"380 ","pages":"Pages 362-374"},"PeriodicalIF":10.5,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142991653","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
A step forwarded on the in vitro and in vivo validation of rifabutin-loaded liposomes for the management of highly virulent MRSA infections
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-08 DOI: 10.1016/j.jconrel.2025.01.083
Mariana P. Coelho , Jacinta O. Pinho , Sandra N. Pinto , Maria Manuela Gaspar
Staphylococcus aureus (S. aureus) infections, especially methicillin resistant (MRSA), constitute an alarming public health issue due to its association with high mortality, morbidity, and hospitalization costs. The increasing antibiotic resistance and biofilm-associated infections of MRSA prompted the discovery of novel and more effective therapeutic strategies. Our team has been working on alternative therapies against S. aureus infections. For this, we have been repurposing an existing antibacterial drug, rifabutin (RFB), through its association to a nanotechnological platform, liposomes, aiming to promote a preferential targeting to infected sites and maximizing its potential antibacterial effect. The therapeutic potential of RFB formulations against a MRSA commercial strain (MRSA ATCC®-33592), either in planktonic or biofilm forms, was assessed. RFB displayed higher antibacterial effects towards biofilm than vancomycin (VCM), the gold standard treatment against MRSA infections, with MBIC50 values of 103 and > 800 μg/mL, respectively. Moreover, the antimicrobial effect of RFB-loaded liposomes demonstrated to be lipid composition-dependent based on MIC50 and MBIC50 values, which was also confirmed by confocal laser scanning microscopy. These studies supported that for positively charged RFB liposomes an electrostatic interaction at biofilm surface occurs without internalization. On the other hand, for RFB-loaded liposomes with neutral surface charge a high internalization within the biofilm was observed. Moreover, this RFB liposomal formulation has also demonstrated to be stable in human plasma, as more than 83 % of RFB was still associated to liposomes 24 h after incubation at 37 °C. The proof of concept of RFB formulations was assessed in MRSA systemic murine models of infection. Therapeutic effect and survival rates were evaluated for animals induced and treated with RFB in free and liposomal forms and compared with negative and positive controls. For the lower infection murine model, 100 % survival was achieved for all groups under study. However, in a higher infection model only for the group of animals treated with RFB incorporated in liposomes a 100 % survival was attained. In terms of bacterial burden, RFB formulations exhibited lower levels when compared to VCM, even using a lower therapeutic dose: 20 vs 40 mg/kg of body weight, respectively. Overall, RFB constitutes an alternative and effective therapeutic strategy towards MRSA infections, being this effect potentiated through its association to a lipid nanoplatform.
{"title":"A step forwarded on the in vitro and in vivo validation of rifabutin-loaded liposomes for the management of highly virulent MRSA infections","authors":"Mariana P. Coelho ,&nbsp;Jacinta O. Pinho ,&nbsp;Sandra N. Pinto ,&nbsp;Maria Manuela Gaspar","doi":"10.1016/j.jconrel.2025.01.083","DOIUrl":"10.1016/j.jconrel.2025.01.083","url":null,"abstract":"<div><div><em>Staphylococcus aureus</em> (<em>S. aureus</em>) infections, especially methicillin resistant (MRSA), constitute an alarming public health issue due to its association with high mortality, morbidity, and hospitalization costs. The increasing antibiotic resistance and biofilm-associated infections of MRSA prompted the discovery of novel and more effective therapeutic strategies. Our team has been working on alternative therapies against <em>S. aureus</em> infections. For this, we have been repurposing an existing antibacterial drug, rifabutin (RFB), through its association to a nanotechnological platform, liposomes, aiming to promote a preferential targeting to infected sites and maximizing its potential antibacterial effect. The therapeutic potential of RFB formulations against a MRSA commercial strain (MRSA ATCC®-33592), either in planktonic or biofilm forms, was assessed. RFB displayed higher antibacterial effects towards biofilm than vancomycin (VCM), the gold standard treatment against MRSA infections, with MBIC<sub>50</sub> values of 103 and &gt; 800 μg/mL, respectively. Moreover, the antimicrobial effect of RFB-loaded liposomes demonstrated to be lipid composition-dependent based on MIC<sub>50</sub> and MBIC<sub>50</sub> values, which was also confirmed by confocal laser scanning microscopy. These studies supported that for positively charged RFB liposomes an electrostatic interaction at biofilm surface occurs without internalization. On the other hand, for RFB-loaded liposomes with neutral surface charge a high internalization within the biofilm was observed. Moreover, this RFB liposomal formulation has also demonstrated to be stable in human plasma, as more than 83 % of RFB was still associated to liposomes 24 h after incubation at 37 °C. The proof of concept of RFB formulations was assessed in MRSA systemic murine models of infection. Therapeutic effect and survival rates were evaluated for animals induced and treated with RFB in free and liposomal forms and compared with negative and positive controls. For the lower infection murine model, 100 % survival was achieved for all groups under study. However, in a higher infection model only for the group of animals treated with RFB incorporated in liposomes a 100 % survival was attained. In terms of bacterial burden, RFB formulations exhibited lower levels when compared to VCM, even using a lower therapeutic dose: 20 <em>vs</em> 40 mg/kg of body weight, respectively. Overall, RFB constitutes an alternative and effective therapeutic strategy towards MRSA infections, being this effect potentiated through its association to a lipid nanoplatform.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"380 ","pages":"Pages 348-361"},"PeriodicalIF":10.5,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143122876","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
BioJect: An in vitro platform to explore release dynamics of peptides in subcutaneous drug delivery.
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-07 DOI: 10.1016/j.jconrel.2025.02.013
David Li, Qin Qiuhua, Ayca Altay Benetti, Burcu Uner, Lyes Kahouadji, Matthias G Wacker

Predicting the release performance of subcutaneous (SC) drug formulations is challenging due to the complex interplay between physicochemical properties and the physiological microenvironment, which includes the extracellular matrix (ECM), fluid composition, and fluid availability, factors that collectively influence bioavailability and absorption rates. The ECM often acts as a bandpass filter modulated by local ion and protein content. In this study, we introduce the BioJect cell, a modern release test method based on the compendial flow-through cell, integrating a perfusion system with customizable biomatrix components. We systematically investigated the release mechanisms of four insulin formulations: regular human insulin, insulin aspart, insulin glulisine, and Neutral Protamine Hagedorn (NPH) insulin. A modified simulated subcutaneous interstitial fluid (mSSIF) comprising multiple components of the SC physiological environment was employed. It incorporates important ions and proteins (138.5 mM sodium, 10 mM potassium, 1.8 mM calcium, 0.8 mM magnesium, 111.3 mM chloride, 28 mM bicarbonate, 0.5 mM sulfate, 5 mM acetate, 4.2 mM phosphate, 30 g/L total protein added as bovine serum albumin). Our release test method discriminated the tested formulations under varying biorelevant conditions, demonstrating its biopredictive capabilities. Notably, we discovered a previously undocumented albumin binding affecting the release rate of insulin glulisine, likely occurring in the low-shear environment of SC tissue only. Additionally, the inclusion of biorelevant components like hyaluronic acid and collagen into the biomatrix of the BioJect cell provided profound insights into potential absorption and release mechanisms, supported by two in vitro-in vivo relationships (level C and level A). The BioJect cell represents a significant advancement in simulating the SC environment for drug release testing. Our findings highlight the importance of considering protein binding and ECM components in predicting drug absorption, offering a promising tool for the development and optimization of SC formulations.

{"title":"BioJect: An in vitro platform to explore release dynamics of peptides in subcutaneous drug delivery.","authors":"David Li, Qin Qiuhua, Ayca Altay Benetti, Burcu Uner, Lyes Kahouadji, Matthias G Wacker","doi":"10.1016/j.jconrel.2025.02.013","DOIUrl":"https://doi.org/10.1016/j.jconrel.2025.02.013","url":null,"abstract":"<p><p>Predicting the release performance of subcutaneous (SC) drug formulations is challenging due to the complex interplay between physicochemical properties and the physiological microenvironment, which includes the extracellular matrix (ECM), fluid composition, and fluid availability, factors that collectively influence bioavailability and absorption rates. The ECM often acts as a bandpass filter modulated by local ion and protein content. In this study, we introduce the BioJect cell, a modern release test method based on the compendial flow-through cell, integrating a perfusion system with customizable biomatrix components. We systematically investigated the release mechanisms of four insulin formulations: regular human insulin, insulin aspart, insulin glulisine, and Neutral Protamine Hagedorn (NPH) insulin. A modified simulated subcutaneous interstitial fluid (mSSIF) comprising multiple components of the SC physiological environment was employed. It incorporates important ions and proteins (138.5 mM sodium, 10 mM potassium, 1.8 mM calcium, 0.8 mM magnesium, 111.3 mM chloride, 28 mM bicarbonate, 0.5 mM sulfate, 5 mM acetate, 4.2 mM phosphate, 30 g/L total protein added as bovine serum albumin). Our release test method discriminated the tested formulations under varying biorelevant conditions, demonstrating its biopredictive capabilities. Notably, we discovered a previously undocumented albumin binding affecting the release rate of insulin glulisine, likely occurring in the low-shear environment of SC tissue only. Additionally, the inclusion of biorelevant components like hyaluronic acid and collagen into the biomatrix of the BioJect cell provided profound insights into potential absorption and release mechanisms, supported by two in vitro-in vivo relationships (level C and level A). The BioJect cell represents a significant advancement in simulating the SC environment for drug release testing. Our findings highlight the importance of considering protein binding and ECM components in predicting drug absorption, offering a promising tool for the development and optimization of SC formulations.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":""},"PeriodicalIF":10.5,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143382385","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
Corrigendum to ‘Investigating the delivery of PD-L1-targeted immunoliposomes in a dynamic cervical cancer-on-a-chip model’ [Journal of Controlled Release Volume 379, 10 March 2025, Pages 236–250]
IF 10.8 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-07 DOI: 10.1016/j.jconrel.2025.01.089
No Abstract
{"title":"Corrigendum to ‘Investigating the delivery of PD-L1-targeted immunoliposomes in a dynamic cervical cancer-on-a-chip model’ [Journal of Controlled Release Volume 379, 10 March 2025, Pages 236–250]","authors":"","doi":"10.1016/j.jconrel.2025.01.089","DOIUrl":"https://doi.org/10.1016/j.jconrel.2025.01.089","url":null,"abstract":"No Abstract","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"85 1","pages":""},"PeriodicalIF":10.8,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143258063","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
Noninvasive assessment of a bioconductive patch for treating atrial fibrillation with magnetic resonance imaging
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-07 DOI: 10.1016/j.jconrel.2025.01.092
Adrian Tabora Dychiao , Ting-Hsuan Lu , Sheng-Yao Peng , Cheng Fan , Siyang Song , Chongyu Zhang , Minyao Wang , Sophia Shi , Jun Wu , Shu-Hong Li , Yen Chang , Hsing-Wen Sung , Ren-Ke Li
Electrical and structural remodeling disrupt atrial electrical conduction, leading to atrial fibrillation (AF). Epicardially delivered conductive biomaterial patches can effectively transmit electrical signals and potentially diminish AF. However, given the progressive nature of AF development, continuous and noninvasive monitoring is essential for assessing the therapeutic efficacy of these patches over time. In this study, superparamagnetic iron oxide nanoparticles (SPIO NPs) are synthesized and used to label a bio-conductive patch made of poly-3-amino-4-methoxybenzoic acid (PAMB) conjugated to gelatin (PAMBG-NP). Incorporating SPIO NPs does not alter the mechanical, electrical, or biocompatible properties of PAMBG. PAMBG-NP restores conduction velocity, suppresses rotor generation, and prevents re-entry currents, thereby relieving AF burden in an in vitro pacing model. In vivo, a bell-shaped PAMBG-NP patch is applied to the right and left atria of KCNE1 knockout mice. Compared to its Gelatin-NP counterpart, PAMBG-NP significantly reduces AF duration and enhances post-AF recovery over a 60-day period. Furthermore, magnetic resonance imaging indicates that PAMBG-NP degrades more slowly than Gelatin-NP, along with having a reduced incidence of AF in PAMBG-NP-treated animals. Therefore, incorporating SPIO NPs into PAMBG enables real-time, in vivo monitoring, potentially facilitating the noninvasive evaluation of its therapeutic efficacy.
{"title":"Noninvasive assessment of a bioconductive patch for treating atrial fibrillation with magnetic resonance imaging","authors":"Adrian Tabora Dychiao ,&nbsp;Ting-Hsuan Lu ,&nbsp;Sheng-Yao Peng ,&nbsp;Cheng Fan ,&nbsp;Siyang Song ,&nbsp;Chongyu Zhang ,&nbsp;Minyao Wang ,&nbsp;Sophia Shi ,&nbsp;Jun Wu ,&nbsp;Shu-Hong Li ,&nbsp;Yen Chang ,&nbsp;Hsing-Wen Sung ,&nbsp;Ren-Ke Li","doi":"10.1016/j.jconrel.2025.01.092","DOIUrl":"10.1016/j.jconrel.2025.01.092","url":null,"abstract":"<div><div>Electrical and structural remodeling disrupt atrial electrical conduction, leading to atrial fibrillation (AF). Epicardially delivered conductive biomaterial patches can effectively transmit electrical signals and potentially diminish AF. However, given the progressive nature of AF development, continuous and noninvasive monitoring is essential for assessing the therapeutic efficacy of these patches over time. In this study, superparamagnetic iron oxide nanoparticles (SPIO NPs) are synthesized and used to label a bio-conductive patch made of poly-3-amino-4-methoxybenzoic acid (PAMB) conjugated to gelatin (PAMBG-NP). Incorporating SPIO NPs does not alter the mechanical, electrical, or biocompatible properties of PAMBG. PAMBG-NP restores conduction velocity, suppresses rotor generation, and prevents re-entry currents, thereby relieving AF burden in an <em>in vitro</em> pacing model. <em>In vivo</em>, a bell-shaped PAMBG-NP patch is applied to the right and left atria of KCNE1 knockout mice. Compared to its Gelatin-NP counterpart, PAMBG-NP significantly reduces AF duration and enhances post-AF recovery over a 60-day period. Furthermore, magnetic resonance imaging indicates that PAMBG-NP degrades more slowly than Gelatin-NP, along with having a reduced incidence of AF in PAMBG-NP-treated animals. Therefore, incorporating SPIO NPs into PAMBG enables real-time, <em>in vivo</em> monitoring, potentially facilitating the noninvasive evaluation of its therapeutic efficacy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"380 ","pages":"Pages 317-329"},"PeriodicalIF":10.5,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143255745","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
Prebiotics empower probiotics with gastrointestinal stress resistance for colon-targeted release to synergistically alleviate colitis
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-07 DOI: 10.1016/j.jconrel.2025.01.059
Haihua Ji, Xiaochen Yan, Li Zhang, Lin Yang, Pengcheng Xie, Fengying Gu, Shuigen Bian, Hao Wan, Shaoping Nie
Oral administration of probiotics holds promise for alleviating ulcerative colitis (UC), yet their efficacy is inevitably compromised by the hostile gastrointestinal (GI) environment. Here, we devised a strategy by coating β-glucan (GN) prebiotic onto the surface of Lactobacillus plantarum (Lp) probiotic at the single-cell level (Lp@CGN) based on bioorthogonal chemistry in a layer-by-layer manner. This achieved to form a firm, dense, and multifunctional GN-based “armor” with advances of superior protective properties, colon-targeted degradation, and prebiotic benefits. Under the protection of the prebiotic-based “armor”, Lp@CGN exhibited a notable 276-fold increase in the survival rate compared to naïve Lp after exposure to whole GI conditions. Upon reaching the colon, the “armor” was metabolized into short-chain fatty acids (SCFAs) by gut microbiota, facilitating the timely release of Lp within colon, thereby achieving a synergistic treatment effect due to sustained SCFAs generation and Lp liberation. As a result, oral administration of Lp@CGN efficiently realized the alleviation of UC in both preventative and therapeutic models through restoring intestinal mucosal barriers, positively regulating inflammatory cytokines, renovating the dysbiosis of gut microbiota, and promoting SCFAs production. In sum, our strategy marks the reconstruction of probiotics with chemical tools, offering useful insights into powering probiotics for disease treatment.
{"title":"Prebiotics empower probiotics with gastrointestinal stress resistance for colon-targeted release to synergistically alleviate colitis","authors":"Haihua Ji,&nbsp;Xiaochen Yan,&nbsp;Li Zhang,&nbsp;Lin Yang,&nbsp;Pengcheng Xie,&nbsp;Fengying Gu,&nbsp;Shuigen Bian,&nbsp;Hao Wan,&nbsp;Shaoping Nie","doi":"10.1016/j.jconrel.2025.01.059","DOIUrl":"10.1016/j.jconrel.2025.01.059","url":null,"abstract":"<div><div>Oral administration of probiotics holds promise for alleviating ulcerative colitis (UC), yet their efficacy is inevitably compromised by the hostile gastrointestinal (GI) environment. Here, we devised a strategy by coating β-glucan (GN) prebiotic onto the surface of <em>Lactobacillus plantarum</em> (<em>Lp</em>) probiotic at the single-cell level (<em>Lp</em>@CGN) based on bioorthogonal chemistry in a layer-by-layer manner. This achieved to form a firm, dense, and multifunctional GN-based “armor” with advances of superior protective properties, colon-targeted degradation, and prebiotic benefits. Under the protection of the prebiotic-based “armor”, <em>Lp</em>@CGN exhibited a notable 276-fold increase in the survival rate compared to naïve <em>Lp</em> after exposure to whole GI conditions. Upon reaching the colon, the “armor” was metabolized into short-chain fatty acids (SCFAs) by gut microbiota, facilitating the timely release of <em>Lp</em> within colon, thereby achieving a synergistic treatment effect due to sustained SCFAs generation and <em>Lp</em> liberation. As a result, oral administration of <em>Lp</em>@CGN efficiently realized the alleviation of UC in both preventative and therapeutic models through restoring intestinal mucosal barriers, positively regulating inflammatory cytokines, renovating the dysbiosis of gut microbiota, and promoting SCFAs production. In sum, our strategy marks the reconstruction of probiotics with chemical tools, offering useful insights into powering probiotics for disease treatment.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"380 ","pages":"Pages 297-316"},"PeriodicalIF":10.5,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143122968","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
Restoring tumor antigenicity activates the “bystander” T cell immune cycle
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-06 DOI: 10.1016/j.jconrel.2025.01.094
Yifan Yang , Qiumin Yu , Haoyu Zhang , Yuchen Liu , Hexuan Wang , Ningyi Yang , Yulian Shi , Wanli Zhang , Zijie Wu , Shitong Huang , Wenbin Xie , Ran Duan , Qiuli Mao , Xupeiyao Shi , Zheng Gao , Xiaoning Wang , Hanlin Guo , Lingxiao Chen , Yi Han , Ximing Li , Xiangdong Gao
Tumor-specific T cells play a crucial role in tumor immunity. However, these cells are often scarce and functionally exhausted within the tumor microenvironment (TME), leading to the limited efficacy of immunotherapy in many cancer patients. In contrast, increasing evidence suggests that the TME is rich in “bystander” T cells (TBYS), most of which are virus-specific and unrelated to the tumor. These TBYS cells retain functional memory characteristics and the potential to kill tumor cells. To utilize TBYS cells in the TME for tumor elimination, we designed an intracellular delivery system, ASCP, encoding a TBYS epitope to redirect tumor cell antigen specificity toward pre-existing TBYS cells, resulting in effective tumor inhibition in multiple preclinical models. The ASCP-antigen peptide strategy restores the antigenicity of tumor cells and induces epitope spreading of tumor antigens, thereby eliciting more diverse tumor-specific T cell responses. Remarkably, this strategy incorporates MHC-II epitopes containing unnatural amino acids (p-nitrophenylalanine, termed NiraTh), which stimulate CD4+ T cell-mediated immunity and assist CD8+ T cells in clearing tumors. Overall, the ASCP-mediated tumor antigen reprogramming strategy provides important insights for cancer immunotherapy in populations with a history of common viral infections.
{"title":"Restoring tumor antigenicity activates the “bystander” T cell immune cycle","authors":"Yifan Yang ,&nbsp;Qiumin Yu ,&nbsp;Haoyu Zhang ,&nbsp;Yuchen Liu ,&nbsp;Hexuan Wang ,&nbsp;Ningyi Yang ,&nbsp;Yulian Shi ,&nbsp;Wanli Zhang ,&nbsp;Zijie Wu ,&nbsp;Shitong Huang ,&nbsp;Wenbin Xie ,&nbsp;Ran Duan ,&nbsp;Qiuli Mao ,&nbsp;Xupeiyao Shi ,&nbsp;Zheng Gao ,&nbsp;Xiaoning Wang ,&nbsp;Hanlin Guo ,&nbsp;Lingxiao Chen ,&nbsp;Yi Han ,&nbsp;Ximing Li ,&nbsp;Xiangdong Gao","doi":"10.1016/j.jconrel.2025.01.094","DOIUrl":"10.1016/j.jconrel.2025.01.094","url":null,"abstract":"<div><div>Tumor-specific T cells play a crucial role in tumor immunity. However, these cells are often scarce and functionally exhausted within the tumor microenvironment (TME), leading to the limited efficacy of immunotherapy in many cancer patients. In contrast, increasing evidence suggests that the TME is rich in “bystander” T cells (T<sub>BYS</sub>), most of which are virus-specific and unrelated to the tumor. These T<sub>BYS</sub> cells retain functional memory characteristics and the potential to kill tumor cells. To utilize T<sub>BYS</sub> cells in the TME for tumor elimination, we designed an intracellular delivery system, ASCP, encoding a T<sub>BYS</sub> epitope to redirect tumor cell antigen specificity toward pre-existing T<sub>BYS</sub> cells, resulting in effective tumor inhibition in multiple preclinical models. The ASCP-antigen peptide strategy restores the antigenicity of tumor cells and induces epitope spreading of tumor antigens, thereby eliciting more diverse tumor-specific T cell responses. Remarkably, this strategy incorporates MHC-II epitopes containing unnatural amino acids (<em>p</em>-nitrophenylalanine, termed NiraTh), which stimulate CD4<sup>+</sup> T cell-mediated immunity and assist CD8<sup>+</sup> T cells in clearing tumors. Overall, the ASCP-mediated tumor antigen reprogramming strategy provides important insights for cancer immunotherapy in populations with a history of common viral infections.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"380 ","pages":"Pages 256-268"},"PeriodicalIF":10.5,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143080137","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
Research trends in the use of nanobodies for cancer therapy.
IF 10.5 1区 医学 Q1 CHEMISTRY, MULTIDISCIPLINARY Pub Date : 2025-02-06 DOI: 10.1016/j.jconrel.2025.01.045
Jun Hou, Kejiang Du, Jinling Li, Zhenghui Li, Shaorui Cao, Shilin Zhang, Wenxing Huang, Heng Liu, Xiaomei Yang, Shuyang Sun, Shanzhao Mo, Tianyu Qin, Xilei Zhang, Shihua Yin, Xinyu Nie, Xiaoling Lu

Although there are many challenges in using nanobodies for treating various complex tumor diseases, including rapid renal clearance and the complex blood-brain barrier environment, nanobodies have shown great potential due to their high antigen affinity, excellent tumor penetration ability, and favorable safety profile. Since the discovery of the variable domain (VHH) of camelid heavy-chain antibodies in 1993, nanobodies have been progressively applied to various cancer therapy platforms, such as antagonistic drugs and targeting agents for effector domains. In recent years, several nanobody-based drugs, including Caplacizumab, KN-035, and Ozoralizumab, have been approved for clinical use. Among them, KN-035 is used for treating advanced solid tumors, and these advancements have propelled nanobody development to new heights. Currently, nanobodies are being rapidly applied to the treatment of a wide range of diseases, from viral infections to cancer, demonstrating strong advantages in areas such as targeted protein degradation, bioimaging, nanobody-drug conjugation, bispecific T-cell engagers, and vaccine applications. Bibliometric tools, including CiteSpace, HisCite Pro, and Alluvial Generator, were employed to trace the historical development of nanobodies in cancer research. The contributions of authors, countries, and institutions in this field were analyzed, and research hotspots and emerging trends were identified through keyword analysis and influential articles. Future trends were also predicted. This study provides a unique, comprehensive, and objective perspective on the use of nanobodies in tumor research, laying a foundation for future research directions and offering valuable insights for researchers in the field.

{"title":"Research trends in the use of nanobodies for cancer therapy.","authors":"Jun Hou, Kejiang Du, Jinling Li, Zhenghui Li, Shaorui Cao, Shilin Zhang, Wenxing Huang, Heng Liu, Xiaomei Yang, Shuyang Sun, Shanzhao Mo, Tianyu Qin, Xilei Zhang, Shihua Yin, Xinyu Nie, Xiaoling Lu","doi":"10.1016/j.jconrel.2025.01.045","DOIUrl":"https://doi.org/10.1016/j.jconrel.2025.01.045","url":null,"abstract":"<p><p>Although there are many challenges in using nanobodies for treating various complex tumor diseases, including rapid renal clearance and the complex blood-brain barrier environment, nanobodies have shown great potential due to their high antigen affinity, excellent tumor penetration ability, and favorable safety profile. Since the discovery of the variable domain (VHH) of camelid heavy-chain antibodies in 1993, nanobodies have been progressively applied to various cancer therapy platforms, such as antagonistic drugs and targeting agents for effector domains. In recent years, several nanobody-based drugs, including Caplacizumab, KN-035, and Ozoralizumab, have been approved for clinical use. Among them, KN-035 is used for treating advanced solid tumors, and these advancements have propelled nanobody development to new heights. Currently, nanobodies are being rapidly applied to the treatment of a wide range of diseases, from viral infections to cancer, demonstrating strong advantages in areas such as targeted protein degradation, bioimaging, nanobody-drug conjugation, bispecific T-cell engagers, and vaccine applications. Bibliometric tools, including CiteSpace, HisCite Pro, and Alluvial Generator, were employed to trace the historical development of nanobodies in cancer research. The contributions of authors, countries, and institutions in this field were analyzed, and research hotspots and emerging trends were identified through keyword analysis and influential articles. Future trends were also predicted. This study provides a unique, comprehensive, and objective perspective on the use of nanobodies in tumor research, laying a foundation for future research directions and offering valuable insights for researchers in the field.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":""},"PeriodicalIF":10.5,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143374220","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
Journal of Controlled Release
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:481959085
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