{"title":"Allan S. Hoffman's leadership in drug delivery: A historical perspective","authors":"Buddy D. Ratner","doi":"10.1016/j.jconrel.2025.114007","DOIUrl":"10.1016/j.jconrel.2025.114007","url":null,"abstract":"","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 114007"},"PeriodicalIF":10.5,"publicationDate":"2025-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144566562","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}
Zimai Liu , Xiaoxi Wang , Xueqin Zhu , Tiantian Zhang , Zonghong He , Zixian Wu , Meiyi Liu , Kai Li , Yuanyuan Wei , Hui Liu , Jiao Lu , Qianxi Lu , Pingping Zhu , Yongchao Wang , Zhenzhen Chen
{"title":"Enhancing antigen presentation in cancer stem cells via peptide-based nanoparticles for effective immunotherapy","authors":"Zimai Liu , Xiaoxi Wang , Xueqin Zhu , Tiantian Zhang , Zonghong He , Zixian Wu , Meiyi Liu , Kai Li , Yuanyuan Wei , Hui Liu , Jiao Lu , Qianxi Lu , Pingping Zhu , Yongchao Wang , Zhenzhen Chen","doi":"10.1016/j.jconrel.2025.114001","DOIUrl":"10.1016/j.jconrel.2025.114001","url":null,"abstract":"<div><div>Cancer stem cells (CSCs) are a subpopulation of cancer cells with the capacity for self-renewal and therapy resistance. The downregulation of major histocompatibility complex class I (MHC-I) molecules, which affects antigen presentation and is a crucial immune evasion mechanism utilized by CSCs, results in their resistance to immunotherapy. Here, we developed an amphiphilic peptide-based nanoparticle, named Smac-D1@Taz, with the PD-L1 antagonist <sup>D</sup>PPA-1 constituting the hydrophilic segment and the Smac peptide, known to enhance radiosensitivity, forming the hydrophobic portion. Tazemetostat (Taz), a small molecule capable of upregulating MHC-I expression in CSCs, was encapsulated in the nanoparticle core during self-assembly. Upon intravenous administration, Smac-D1@Taz exhibited preferential accumulation at the tumor site. The matrix metalloproteinase-2 (MMP2) overexpressed in the tumor environment triggers the disassembly of the nanoparticle. The released Smac peptide enhances the radiosensitivity of tumor cells, while the PD-L1 antagonist <sup>D</sup>PPA-1 restores the function of T cells. Meanwhile, Taz upregulates MHC-I expression and enhances antigen presentation on CSCs, promoting their recognition and elimination by T cells. The results showed that Smac-D1@Taz significantly inhibited tumor progression and reduced the risk of postoperative recurrence and metastasis. This study offers an effective strategy to enhance the therapeutic efficacy of immunotherapy against CSCs, leading to improved cancer treatment outcomes.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 114001"},"PeriodicalIF":10.5,"publicationDate":"2025-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144566561","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}
Jam-Eon Park , Soo-Ji Kang , Jun-Seong Ahn , Seung-Hyeon Choi , Jung-Sook Lee , Ji-Sun Kim , Seung-Hwan Park
{"title":"Optimized bacterial delivery approaches for precision immunotherapy in breast cancer models","authors":"Jam-Eon Park , Soo-Ji Kang , Jun-Seong Ahn , Seung-Hyeon Choi , Jung-Sook Lee , Ji-Sun Kim , Seung-Hwan Park","doi":"10.1016/j.jconrel.2025.114004","DOIUrl":"10.1016/j.jconrel.2025.114004","url":null,"abstract":"<div><div>Bacteria-mediated cancer therapy is an innovative approach that exploits the tumor-targeting ability of bacteria to deliver anti-cancer drugs directly to tumors. Cytolysin A (ClyA), a bacterial pore-forming toxin, has demonstrated therapeutic efficacy in colorectal cancer but has limited effectiveness in breast cancer. To address this limitation, we engineered an attenuated <em>Salmonella</em> strain to express <em>Clostridium perfringens</em> enterotoxin (CPE), which selectively targets CLDN-4, a tight junction protein overexpressed in breast cancer, thereby minimizing off-target effects. In a 4T1 breast tumor mouse model, CPE-secreting bacteria demonstrated significantly greater therapeutic efficacy than ClyA-secreting bacteria. Mechanistic investigations revealed that ClyA and CPE induced distinct patterns of immune cell infiltration depending on tumor type. In 4T1 tumors, CPE significantly increased the infiltration of CD4<sup>+</sup> T cells, CD8<sup>+</sup> T cells, and NK cells, while reducing neutrophil infiltration. In contrast, ClyA promoted immune cell infiltration in CT26 tumors but had negligible effects in 4T1 tumors. Furthermore, CPE treatment markedly reduced granulocyte-colony stimulating factor (G-CSF) expression in 4T1 tumors, a key regulator of neutrophil recruitment, tumor growth, and chemotherapy resistance. Our findings demonstrate that CPE-secreting bacteria exert superior therapeutic efficacy through two synergistic mechanisms: (1) direct tumor cell lysis <em>via</em> pore formation and apoptosis induction, and (2) modulation of the tumor immune microenvironment by enhancing tumor-infiltrating lymphocytes and suppressing neutrophil-associated tumor progression. These results highlight the importance of tailoring bacteria-mediated cancer therapy to tumor specific molecular characteristics to maximize therapeutic efficacy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 114004"},"PeriodicalIF":10.5,"publicationDate":"2025-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565821","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}
Chang Geun Kim , Mincheol Jang , Chanhee Oh , Ok Hwa Jeon , Byeong Hyeon Choi , Kyungsu Kim , Ji-Ho Park , Hyun Koo Kim
{"title":"Pulmonary surfactant-based pirfenidone-loaded nanovesicles for inhalation therapy of idiopathic pulmonary fibrosis","authors":"Chang Geun Kim , Mincheol Jang , Chanhee Oh , Ok Hwa Jeon , Byeong Hyeon Choi , Kyungsu Kim , Ji-Ho Park , Hyun Koo Kim","doi":"10.1016/j.jconrel.2025.114005","DOIUrl":"10.1016/j.jconrel.2025.114005","url":null,"abstract":"<div><div>Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal chronic disease. One of the Food and Drug Administration (FDA)-approved therapies for IPF, oral pirfenidone (PFD), has limited clinical applications owing to its systemic side effects. In contrast to oral administration, inhaled therapy offers enhanced therapeutic efficacy at the target organ while minimizing systemic side effects. However, its application has challenges, such as limited drug delivery to the distal lung region and rapid clearance. In this study, we developed pulmonary surfactant (PS)-based PFD-loaded nanovesicles (PFD-PSNVs) for targeted delivery to the lung area and prolonged retention and examined their safety, stability, and antifibrotic efficacy. PFD-PSNVs were prepared using the thin-film hydration and extrusion method. The mean size and zeta potential of PFD-PSNVs were 149.7 ± 10.1 nm and − 31.3 ± 2.3 mV, respectively. An in vitro antifibrotic study showed that PFD-PSNVs inhibited the expression of fibrotic factors such as p-ERK, p-SMAD2/3, and α-SMA proteins on fibroblasts activated by transforming growth factor-β1. When inhaled in mice using a nebulizer, the PFD-PSNVs remained in lung tissues for 24 h, whereas Arikayce, an FDA-approved liposomal formulation for inhalation, was eliminated within 6 h. In a bleomycin sulfate-induced IPF mouse model, inhalation treatment with PFD-PSNVs significantly reduced collagen deposition (76.2 ± 4.1 %, <em>p</em> < 0.01) and α-SMA expression (60.8 ± 3.7 %, <em>p</em> < 0.05) compared with inhalation treatment with the PFD-loaded CtrlNV (PFD-CNVs) formulation and oral administration of PFD. These results indicate that PSNVs have great potential as an inhaled drug delivery system for the treatment of IPF.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 114005"},"PeriodicalIF":10.5,"publicationDate":"2025-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565841","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}
Xin Yu Liu , He Ying Mao , Jun Sheng Hu , Tong Rui Dou , Ben Chi Liu , Chang Xiu Lin , Cheng-Hua Jin , Ming Guan Piao
{"title":"Reactive oxygen species-responsive micelles targeting activated hepatic stellate cells for treating liver fibrosis","authors":"Xin Yu Liu , He Ying Mao , Jun Sheng Hu , Tong Rui Dou , Ben Chi Liu , Chang Xiu Lin , Cheng-Hua Jin , Ming Guan Piao","doi":"10.1016/j.jconrel.2025.113997","DOIUrl":"10.1016/j.jconrel.2025.113997","url":null,"abstract":"<div><div>Effectively mitigating the progression of liver fibrosis is crucial for preventing the advancement of chronic liver diseases to end-stage cirrhosis and hepatocellular carcinoma. Targeted delivery to hepatic stellate cells (HSCs) has demonstrated promising potential in the treatment of liver fibrosis. However, the development of HSC-targeted drug delivery systems faces significant challenges due to the inefficiency of nano-delivery systems in achieving adequate cellular entry and the limited controllability of drug release. Here, we designed a targeted penetrating peptide, cRGDfK-R6, which comprises the integrin αvβ3-targeting peptide segment cRGDfK and the cell-penetrating peptide R6, conjugated to micelles that are cleavable in response to high levels of reactive oxygen species (ROS) within activated HSCs (aHSCs), aiming to achieve sequential delivery that penetrates the cell membrane and specifically releases the payload upon targeting aHSCs. We synthesized an amphiphilic block copolymer linked by thioketal (TK) and successfully conjugated it with cRGDfK-R6, subsequently self-assembling with betulin (Bt) to form stable and biocompatible Bt/cRGDfK-R6-PPMs<sup>TK</sup>. Compared with micelles lacking cell-penetrating action or non-ROS-responsiveness, Bt/cRGDfK-R6-PPMs<sup>TK</sup> demonstrated enhanced drug accumulation in aHSCs and anti-fibrotic activity both <em>in vitro</em> and in fibrotic mouse models. In summary, this work indicates that Bt/cRGDfK-R6-PPMs<sup>TK</sup> provides an efficient and precise innovative platform for drug delivery to aHSCs in treating liver fibrosis.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 113997"},"PeriodicalIF":10.5,"publicationDate":"2025-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144566564","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}
Jiakun Guo , Hujing Tan , Wanting Chen, Yan Wang, Wenhai Lin, Zhiyuan Zhong, Chao Deng
{"title":"Oncolytic microgels with regulated antibody release augment tumor immunotherapy","authors":"Jiakun Guo , Hujing Tan , Wanting Chen, Yan Wang, Wenhai Lin, Zhiyuan Zhong, Chao Deng","doi":"10.1016/j.jconrel.2025.114003","DOIUrl":"10.1016/j.jconrel.2025.114003","url":null,"abstract":"<div><div>The inhibition of immune checkpoints has emerged as a most successful immunotherapy strategy for cancers; however, it bears a modest clinical response rate and certain cases severe systemic adverse reactions. Here, oncolytic microgels (OMG) that possess similar antitumor activity and immune activation to oncolytic peptide LTX-315 and are capable of sustained release of immune checkpoint inhibitors have been developed to potentiate cancer immunotherapy. Of note, antibodies including anti-PD-1, anti-PD-L1, and anti-CTLA-4 all could be quantitatively loaded into OMG while being gradually released over a couple of weeks in vitro and in tumor as well. In the B16F10 melanoma model, a single tumoral injection of anti-PD-1 and anti-CTLA-4-loaded OMG (P1C4@OMG) effectively reversed suppressive tumor microenvironment and enhanced anti-tumor immune response, achieving potent tumor suppression and striking survival benefits. This study heralds oncolytic microgels with regulated antibody release as a safe and unique platform for cancer immunotherapy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 114003"},"PeriodicalIF":10.5,"publicationDate":"2025-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144565819","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}
Jiang Yu , Zewei Tu , Jiali Fan , Kunjian Lei , Zhouqi Meng , Binfan Chen , Zefeng Wang , William Escobar , Jiangbing Zhou
{"title":"Phosphatidylcholine-derived carriers facilitate brain tumor delivery and enhance glioblastoma therapy","authors":"Jiang Yu , Zewei Tu , Jiali Fan , Kunjian Lei , Zhouqi Meng , Binfan Chen , Zefeng Wang , William Escobar , Jiangbing Zhou","doi":"10.1016/j.jconrel.2025.113999","DOIUrl":"10.1016/j.jconrel.2025.113999","url":null,"abstract":"<div><div>Glioblastoma (GBM) is the most common primary brain cancer without effective treatment. The ineffective treatment of GBM can be partially attributed to the existence of the blood-brain barrier (BBB). Lipids, which constitute over half the weight of the brain and play a vital role in brain tumor biology, can be transported to the brain in the form of lysophosphatidylcholines (LPCs) via specific LPC transporters at the BBB. We hypothesize that LPC analogs could be used as carriers for drug delivery to tumors in the brain. To test this hypothesis, we synthesized and screened a collection of LPC analogs, among which LPC analog 3 (A3), featuring a glycerophosphorylcholine (GPC) headgroup and a 15‑carbon tail, exhibited a marked ability to penetrate brain tumors. We characterized A3 as a carrier for drug delivery to brain tumors by using Doxorubicin (Dox) as the therapeutic payload and found that the A3-Dox conjugate with a cathepsin B-cleavable linker has a great ability to accumulate in brain tumors, leading to effective treatment of GBM without inducing significant cytotoxicity. Our study suggests a novel approach to improving the treatment of GBM by enhancing the delivery of therapeutic agents to the brain using A3 as a carrier.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 113999"},"PeriodicalIF":10.5,"publicationDate":"2025-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144566563","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}
Qiongqiong Yao , Fan Zhang , Zhaoying Yang , Yuanzi Sun , Weiping Gao
{"title":"A super-long-acting anti-PD-L1 nanobody fused to elastin-like polypeptide for triple-negative breast cancer immunotherapy","authors":"Qiongqiong Yao , Fan Zhang , Zhaoying Yang , Yuanzi Sun , Weiping Gao","doi":"10.1016/j.jconrel.2025.114002","DOIUrl":"10.1016/j.jconrel.2025.114002","url":null,"abstract":"<div><div>The limited tumor penetration of monoclonal antibodies or the rapid renal clearance of nanobodies hamper their application in cancer immunotherapy. Through genetic engineering, we fuse a thermosensitive elastin-like polypeptide of ELP(V) with a nanobody of KN035 that binds specifically to human programmed cell death protein ligand 1 (PD-L1) to generate a conjugate of KN035-ELP(V) for triple-negative breast cancer (TNBC) therapy. The high affinity of KN035-ELP(V) enables it to efficiently block the interaction of PD1 and PD-L1. A single intraperitoneal administration of KN035-ELP(V) leads to the <em>in situ</em> generation of a sustained-release depot, showing a circulating half-life that is 276.4-fold and 5.1-fold longer than those of KN035 and KN035 fused with fragment crystallizable (KN035-Fc), respectively. Additionally, KN035-ELP(V) outperforms KN035 and KN035-Fc in term of accumulation and penetration in tumor tissue. Compared to KN035 and KN035-Fc treatments, a single injection of KN035-ELP(V) induces a durable and efficient antitumor immunity in TNBC-bearing mice, resulting in improved anti-tumor efficacy with reduced adverse effects. Therefore, KN035-ELP(V) is promising as a super-long-acting anti-PD-L1 nanobody for efficient cancer immune checkpoint blockade therapy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 114002"},"PeriodicalIF":10.5,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144533819","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}
Jeongjin Lee , Seung Woo Choi , Minsung Park , Wooram Um , Seunglee Kwon , Chan Ho Kim , Hyeyeon Joo , Dong Gil You , Jae Ah Lee , Sol Shin , Heegun Kang , Farrokhroo Ghahari , Jaeyun Kim , Jae Hyung Park
{"title":"Hematite-embedded mesoporous nanoparticles for ferroptosis-inducing cancer sonoimmunotherapy","authors":"Jeongjin Lee , Seung Woo Choi , Minsung Park , Wooram Um , Seunglee Kwon , Chan Ho Kim , Hyeyeon Joo , Dong Gil You , Jae Ah Lee , Sol Shin , Heegun Kang , Farrokhroo Ghahari , Jaeyun Kim , Jae Hyung Park","doi":"10.1016/j.jconrel.2025.113990","DOIUrl":"10.1016/j.jconrel.2025.113990","url":null,"abstract":"<div><div>Sonodynamic therapy (SDT) aims to treat cancers by generating reactive oxygen species in response to ultrasound (US). However, the clinical applications of SDT are often constrained due to its limited efficacy in triggering systemic immune responses. Considering this, we developed hematite-embedded PEGylated mesoporous silica nanoparticles (H@PMSNs) as potential sonosensitizers for inducing immunogenic cancer cell death. When B16F10 cells were exposed to H@PMSNs under US irradiation, the intracellular levels of glutathione and glutathione peroxidase 4 were significantly reduced, leading to lipid peroxidation of the cell membrane and triggering ferroptosis. Notably, after their systemic administration into tumor-bearing mice, H@PMSNs effectively inhibited tumor growth under US irradiation by inducing ferroptosis. This process led to the enhanced maturation of dendritic cells and a significant increase in cytotoxic T cells. Consequently, the antitumor efficacy of immune checkpoint blockade was significantly enhanced by H@PMSN-based SDT. The results of this study indicate that H@PMSNs, which induce the immunogenic death of cancer cells through hematite-mediated ferroptosis, could be promising sonosensitizers for cancer immunotherapy.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"385 ","pages":"Article 113990"},"PeriodicalIF":10.5,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144521213","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}