Pub Date : 2026-01-06DOI: 10.1016/j.jconrel.2026.114608
He Wang, Zhao Wang, Yen Leng Pak, Quanxiang Han, Wenxiu Wu, Yurong Guo, Haohao Zhang, Yingying Jing, Zhiping Zhang, Liwei Chen, Xing Gao, Lei Yu, Jibin Song
Reactive oxygen species (ROS)-based cancer treatment leverages their strong oxidizing properties to damage cellular components, inducing apoptosis and eliminating tumours. In comparison with ROS, reactive nitrogen species (RNS) demonstrate enhanced nitrification and oxidation capabilities, which exert more significant damage on tumour cells. Herein, we described a 980-nm laser-responsive prodrug nanosystem (RCLA) consisted of rare earth nanoparticles (RENPs, NaYF4:Yb, 20 %Er@NaYF4:Yb) and Ce-UiO-66-NH2, which enables the controlled release of L-arginine and efficient production of RNS through cascade enzymatic catalysis. After the modulation of doping Er3+ concentrations, the laser-responsive RCLA not only exhibited excellent NIR-II fluorescence imaging, but also possessed Förster energy transfer to enhance cascade enzymatic activity. In addition, RCLA show high-efficient catalase- and oxidase-like activity to induce the generated O2 to convert into ROS. Then, the excessive ROS cleaves the ROS-sensitive thioketal bond in RCLA to release free L-arginine, and further reacts with L-arginine to subsequently generate RNS. Finally, RNA sequencing analysis reveals that RCLA profoundly affects the normal biological functions of tumour cells by inducing oxidative stress and activating apoptosis-related pathways. Combined with the anti-tumour activity of RCLA in vitro and in vivo, this nanozyme system provides an efficient and precise strategy for synergistic cancer therapy.
{"title":"Near-infrared light-switchable unlocking of tumour-specific prodrug nanosystem for synergistic enhancement of reactive nitrogen species-based therapy.","authors":"He Wang, Zhao Wang, Yen Leng Pak, Quanxiang Han, Wenxiu Wu, Yurong Guo, Haohao Zhang, Yingying Jing, Zhiping Zhang, Liwei Chen, Xing Gao, Lei Yu, Jibin Song","doi":"10.1016/j.jconrel.2026.114608","DOIUrl":"https://doi.org/10.1016/j.jconrel.2026.114608","url":null,"abstract":"<p><p>Reactive oxygen species (ROS)-based cancer treatment leverages their strong oxidizing properties to damage cellular components, inducing apoptosis and eliminating tumours. In comparison with ROS, reactive nitrogen species (RNS) demonstrate enhanced nitrification and oxidation capabilities, which exert more significant damage on tumour cells. Herein, we described a 980-nm laser-responsive prodrug nanosystem (RCLA) consisted of rare earth nanoparticles (RENPs, NaYF<sub>4</sub>:Yb, 20 %Er@NaYF<sub>4</sub>:Yb) and Ce-UiO-66-NH<sub>2</sub>, which enables the controlled release of L-arginine and efficient production of RNS through cascade enzymatic catalysis. After the modulation of doping Er<sup>3+</sup> concentrations, the laser-responsive RCLA not only exhibited excellent NIR-II fluorescence imaging, but also possessed Förster energy transfer to enhance cascade enzymatic activity. In addition, RCLA show high-efficient catalase- and oxidase-like activity to induce the generated O<sub>2</sub> to convert into ROS. Then, the excessive ROS cleaves the ROS-sensitive thioketal bond in RCLA to release free L-arginine, and further reacts with L-arginine to subsequently generate RNS. Finally, RNA sequencing analysis reveals that RCLA profoundly affects the normal biological functions of tumour cells by inducing oxidative stress and activating apoptosis-related pathways. Combined with the anti-tumour activity of RCLA in vitro and in vivo, this nanozyme system provides an efficient and precise strategy for synergistic cancer therapy.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":"114608"},"PeriodicalIF":11.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933682","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}
Pub Date : 2026-01-06DOI: 10.1016/j.jconrel.2026.114604
Longxiao Li, Zifan Pei, Qi Wo, Nan Jiang, Fangyi Yu, Xiaofen Zhang, Nailin Yang, Shumin Sun, Jihu Nie, Jie Wu, Zhicheng Liu, Yinqing Pei, Hua Liu, Sheng Wang, Liang Cheng
Gas therapy, an emerging and promising tumor treatment strategy, has garnered increasing research interest. Recent attention has focused on gas signaling molecules due to their unique biological effects and potent immunomodulatory activities. With rapid advances in nanotechnology, diverse gas-generating nanoplatforms have been developed to augment cancer immunotherapy. This review first elucidates the mechanisms by which bioactive gas molecules amplify anti-tumor immunity and outlines design strategies for constructing gas-generating nanoplatforms. Thereafter, we summarize the applications of these nanoplatforms in gas-enhanced tumor immunotherapy, highlighting key bioactive gas signal molecules, including hydrogen sulfide (H2S), nitric oxide (NO), carbon monoxide (CO), and hydrogen (H2). Finally, the biosafety profiles of these systems and prospect future opportunities and challenges are discussed.
{"title":"Nanostrategies for potentiating gas-immunotherapy: From advanced delivery platforms to anti-tumor applications.","authors":"Longxiao Li, Zifan Pei, Qi Wo, Nan Jiang, Fangyi Yu, Xiaofen Zhang, Nailin Yang, Shumin Sun, Jihu Nie, Jie Wu, Zhicheng Liu, Yinqing Pei, Hua Liu, Sheng Wang, Liang Cheng","doi":"10.1016/j.jconrel.2026.114604","DOIUrl":"https://doi.org/10.1016/j.jconrel.2026.114604","url":null,"abstract":"<p><p>Gas therapy, an emerging and promising tumor treatment strategy, has garnered increasing research interest. Recent attention has focused on gas signaling molecules due to their unique biological effects and potent immunomodulatory activities. With rapid advances in nanotechnology, diverse gas-generating nanoplatforms have been developed to augment cancer immunotherapy. This review first elucidates the mechanisms by which bioactive gas molecules amplify anti-tumor immunity and outlines design strategies for constructing gas-generating nanoplatforms. Thereafter, we summarize the applications of these nanoplatforms in gas-enhanced tumor immunotherapy, highlighting key bioactive gas signal molecules, including hydrogen sulfide (H<sub>2</sub>S), nitric oxide (NO), carbon monoxide (CO), and hydrogen (H<sub>2</sub>). Finally, the biosafety profiles of these systems and prospect future opportunities and challenges are discussed.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":"114604"},"PeriodicalIF":11.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933725","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}
Intratracheal administration demonstrates superior therapeutic effectiveness against pulmonary diseases with reduced systemic toxicity, especially for non-small cell lung cancer (NSCLC). However, conventional intratracheally administered drugs struggle to further penetrate into the tumor core from the pulmonary parenchyma due to biological obstacles such as the mucosal barrier and the tumor extracellular matrix. Here, an engineered Salmonella strain named VNPDCX that achieves flagella overexpression and inducible lysis has been developed as an intratracheal delivery vehicle to penetrate biological barriers for intratumoral drug delivery. After intratracheal administration, the enhanced motility and invasive properties of VNPDCX facilitate them to penetrate barriers and colonize orthotopic lung tumors, along with the stimulator of interferon genes (STING) agonist cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) loaded on them. Bacteria remaining in lung tissues are subsequently lysed via the inducible lysis circuit to avoid refractory pneumonia. The combination of STING pathway activation and bacterial overexpressed flagella facilitates tumor-associated macrophage M1 polarization. Additionally, dendritic cells mature, as well as T lymphocytes prime, both of which promote tumor eradication. Therefore, intratracheal motile bacterial carriers are effective in penetrating biological barriers and delivering therapeutic drugs, offering a novel strategy for non-small cell lung cancer immunotherapy.
{"title":"Flagella-overexpressing bacteria with inducible lysis capability delivering cGAMP intratracheally for enhanced anticancer immunotherapy","authors":"Zhiyan Li, Yanjun Lu, Wenliang Ma, Qingwei Zeng, Hanxiao Xu, Lulu Wang, Tao Wang, Jinhui Wu","doi":"10.1016/j.jconrel.2026.114606","DOIUrl":"https://doi.org/10.1016/j.jconrel.2026.114606","url":null,"abstract":"Intratracheal administration demonstrates superior therapeutic effectiveness against pulmonary diseases with reduced systemic toxicity, especially for non-small cell lung cancer (NSCLC). However, conventional intratracheally administered drugs struggle to further penetrate into the tumor core from the pulmonary parenchyma due to biological obstacles such as the mucosal barrier and the tumor extracellular matrix. Here, an engineered <em>Salmonella</em> strain named VNP<sub>DCX</sub> that achieves flagella overexpression and inducible lysis has been developed as an intratracheal delivery vehicle to penetrate biological barriers for intratumoral drug delivery. After intratracheal administration, the enhanced motility and invasive properties of VNP<sub>DCX</sub> facilitate them to penetrate barriers and colonize orthotopic lung tumors, along with the stimulator of interferon genes (STING) agonist cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) loaded on them. Bacteria remaining in lung tissues are subsequently lysed <em>via</em> the inducible lysis circuit to avoid refractory pneumonia. The combination of STING pathway activation and bacterial overexpressed flagella facilitates tumor-associated macrophage M1 polarization. Additionally, dendritic cells mature, as well as T lymphocytes prime, both of which promote tumor eradication. Therefore, intratracheal motile bacterial carriers are effective in penetrating biological barriers and delivering therapeutic drugs, offering a novel strategy for non-small cell lung cancer immunotherapy.","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"40 1","pages":""},"PeriodicalIF":10.8,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145902493","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}
Pub Date : 2026-01-06DOI: 10.1016/j.jconrel.2026.114609
Ana Baião , Flávia Castro , Juliana Viegas , Andreia S. Barros , Sofia Dias , Carla Oliveira , Bruno Sarmento
Colorectal cancer (CRC) remains a leading cause of cancer mortality, with limited therapeutic options in advanced stages. CD44v6, a splice variant overexpressed in CRC, promotes tumor progression and immune evasion, representing a relevant target for selective drug delivery. In this work, irinotecan-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles functionalized with CD44v6 ligands (Iri@NP-v6) were developed to improve irinotecan delivery and therapeutic responses in CRC. Iri@NP-v6 showed controlled physicochemical properties, high drug loading, and sustained release. In vitro, targeted NPs achieved selective uptake and enhanced cytotoxicity in CD44v6+ CRC cell lines. In immune-stromal co-culture spheroids, Iri@NP-v6 combined with anti-PD-L1 reduced viability, promoted Th1-associated cytokines, and limited tumor-supportive MCP-1 production. While NP monotherapy induced mixed inflammatory signals, PD-L1 blockade redirected this response toward a Th1-dominated profile. In immunocompetent mice with MC38 tumors, combination therapy significantly reduced tumor burden and increased CD4+ and CD8+ T cell infiltration without systemic toxicity. Importantly, free irinotecan triggered broad systemic inflammation, whereas NP-based delivery limited systemic cytokine release while maintaining intratumoral immune activation. These findings demonstrate that CD44v6-targeted irinotecan NPs in combination with PD-L1 blockade, reshape the CRC immune microenvironment while reducing systemic inflammation, supporting their potential as a tumor-selective chemo-immunotherapy platform.
{"title":"Targeted nanoparticle delivery of irinotecan enhances tumor response to PD-L1 blockade in colorectal cancer","authors":"Ana Baião , Flávia Castro , Juliana Viegas , Andreia S. Barros , Sofia Dias , Carla Oliveira , Bruno Sarmento","doi":"10.1016/j.jconrel.2026.114609","DOIUrl":"10.1016/j.jconrel.2026.114609","url":null,"abstract":"<div><div>Colorectal cancer (CRC) remains a leading cause of cancer mortality, with limited therapeutic options in advanced stages. CD44v6, a splice variant overexpressed in CRC, promotes tumor progression and immune evasion, representing a relevant target for selective drug delivery. In this work, irinotecan-loaded poly(lactic-<em>co</em>-glycolic acid) (PLGA) nanoparticles functionalized with CD44v6 ligands (Iri@NP-v6) were developed to improve irinotecan delivery and therapeutic responses in CRC. Iri@NP-v6 showed controlled physicochemical properties, high drug loading, and sustained release. <em>In vitro</em>, targeted NPs achieved selective uptake and enhanced cytotoxicity in CD44v6<sup>+</sup> CRC cell lines. In immune-stromal co-culture spheroids, Iri@NP-v6 combined with anti-PD-L1 reduced viability, promoted Th1-associated cytokines, and limited tumor-supportive MCP-1 production. While NP monotherapy induced mixed inflammatory signals, PD-L1 blockade redirected this response toward a Th1-dominated profile. In immunocompetent mice with MC38 tumors, combination therapy significantly reduced tumor burden and increased CD4<sup>+</sup> and CD8<sup>+</sup> T cell infiltration without systemic toxicity. Importantly, free irinotecan triggered broad systemic inflammation, whereas NP-based delivery limited systemic cytokine release while maintaining intratumoral immune activation. These findings demonstrate that CD44v6-targeted irinotecan NPs in combination with PD-L1 blockade, reshape the CRC immune microenvironment while reducing systemic inflammation, supporting their potential as a tumor-selective chemo-immunotherapy platform.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"391 ","pages":"Article 114609"},"PeriodicalIF":11.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145908107","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}
Pub Date : 2026-01-06DOI: 10.1016/j.jconrel.2026.114615
Hao Yang, Yanling Kuang, Lamei Wang, Huimin Zhang, Rui Zhang, Yanfei Dai, Javier A Villafuerte Gálvez, Junhu Yao, Shimin Liu, Xinhua Chen, Yangchun Cao
Clostridioides difficile infection (CDI) is strongly linked to disruptions in gut microbial homeostasis. Although conventional antibiotics effectively inhibit C. difficile proliferation, they frequently fail to reestablish microbial equilibrium, thereby constraining therapeutic outcomes. Probiotic therapy seeks to modulate gut microecology by replenishing beneficial microorganisms; however, its clinical efficacy is hindered by poor gastrointestinal survivability, limited colon-targeted engraftment, and inadequate suppression of C. difficile virulence determinants. To address these challenges, we developed a polysaccharide-based hydrogel platform (HF), named BA@HF-PDAT, designed for the synergistic co-delivery of Bifidobacterium adolescentis (BA) and polydopamine-thymol nanoparticles (PDA-TH NPs). HF-mediated encapsulation substantially enhances probiotic viability, facilitates colon- and inflammation-targeted controlled release, and promotes bacterial engraftment. Concurrently, PDA-TH effectively inhibits C. difficile virulence determinants, synergistically amplifying probiotic therapeutic potential. In vitro and in vivo analyses reveal that BA@HF-PDAT facilitates intestinal repair and mitigates apoptosis through Wnt/β-catenin signaling, exerts potent anti-inflammatory and antioxidant activities, and restores gut microbial composition and metabolic function, thereby effectively ameliorating CDI-induced colitis in murine models. Comparative studies further indicate that BA@HF-PDAT achieves superior therapeutic efficacy compared with vancomycin treatment. Collectively, this integrated co-delivery platform constitutes a promising and translational therapeutic strategy for CDI management.
{"title":"A multifunctional probiotic co-delivery platform for the treatment of Clostridium difficile infection.","authors":"Hao Yang, Yanling Kuang, Lamei Wang, Huimin Zhang, Rui Zhang, Yanfei Dai, Javier A Villafuerte Gálvez, Junhu Yao, Shimin Liu, Xinhua Chen, Yangchun Cao","doi":"10.1016/j.jconrel.2026.114615","DOIUrl":"https://doi.org/10.1016/j.jconrel.2026.114615","url":null,"abstract":"<p><p>Clostridioides difficile infection (CDI) is strongly linked to disruptions in gut microbial homeostasis. Although conventional antibiotics effectively inhibit C. difficile proliferation, they frequently fail to reestablish microbial equilibrium, thereby constraining therapeutic outcomes. Probiotic therapy seeks to modulate gut microecology by replenishing beneficial microorganisms; however, its clinical efficacy is hindered by poor gastrointestinal survivability, limited colon-targeted engraftment, and inadequate suppression of C. difficile virulence determinants. To address these challenges, we developed a polysaccharide-based hydrogel platform (HF), named BA@HF-PDAT, designed for the synergistic co-delivery of Bifidobacterium adolescentis (BA) and polydopamine-thymol nanoparticles (PDA-TH NPs). HF-mediated encapsulation substantially enhances probiotic viability, facilitates colon- and inflammation-targeted controlled release, and promotes bacterial engraftment. Concurrently, PDA-TH effectively inhibits C. difficile virulence determinants, synergistically amplifying probiotic therapeutic potential. In vitro and in vivo analyses reveal that BA@HF-PDAT facilitates intestinal repair and mitigates apoptosis through Wnt/β-catenin signaling, exerts potent anti-inflammatory and antioxidant activities, and restores gut microbial composition and metabolic function, thereby effectively ameliorating CDI-induced colitis in murine models. Comparative studies further indicate that BA@HF-PDAT achieves superior therapeutic efficacy compared with vancomycin treatment. Collectively, this integrated co-delivery platform constitutes a promising and translational therapeutic strategy for CDI management.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":"114615"},"PeriodicalIF":11.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933593","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}
Pub Date : 2026-01-06DOI: 10.1016/j.jconrel.2026.114603
Yao Gong, Xiaoying Zhang, Wenlong Ren, Jian Peng, Miaoshu Liu, Lanxin Jiang, Caixia Pei, Xingping Hu, Jie Xu, Ting Zhang, Fengxia Gao, Siqiao Li, Li Du, Wei Cheng
Despite the crucial role of natural killer (NK) cells in initiating antitumor immune circuits, their efficacy is limited by tumor immune evasion mechanisms, including low surface density or proteolytic shedding of activating ligands and immunosuppression. Here, we constructed a bioorthogonal truncated NKG2D (natural killer group 2, member D) ligand-based nano-igniter (ZIL15-D-trMULT1) to synchronously enhance NK cell recognition and function, thereby igniting antitumor immune circuits. This system incorporates a truncated NKG2D ligand (trMULT1) lacking the cleavable α3 domain while retaining the NKG2D-binding α1/α2 domains, conjugated to dibenzocyclooctyne (DBCO) for bioorthogonal tagging of azide-modified tumor cells. Concurrently, interleukin-15 (IL-15) was encapsulated within zeolitic imidazolate frameworks (ZIF-8) for responsive release, sustaining NK cell function and upregulating NKG2D receptor expression. This synergistic design created a positive feedback loop for NK cell recognition and activation. Consequently, this strategy suppressed multiple tumor types and improved the immune microenvironment. Mechanistically, both innate and adaptive immunity were mobilized by orchestrating crosstalk between dendritic cells and T cells, facilitated by NK cell-derived chemokine secretion. In combination with anti-PD-1 antibody, ZIL15-D-trMULT1 induced durable immune memory, restraining distal tumor growth and lung metastasis. Our work unveils a self- sustaining immune circuit triggered by NK cells through upregulating the un-cleavable NKG2D ligand density, providing a robust strategy against solid tumors.
尽管自然杀伤(NK)细胞在启动抗肿瘤免疫回路中起着至关重要的作用,但其功效受到肿瘤免疫逃避机制的限制,包括低表面密度或激活配体的蛋白水解脱落和免疫抑制。本研究构建了一个生物正交截断的NKG2D (natural killer group 2, member D)配体纳米点燃剂ZIL15-D-trMULT1,同步增强NK细胞的识别和功能,从而点燃抗肿瘤免疫回路。该系统包含一个截断的NKG2D配体(trMULT1),缺少可切割的α3结构域,同时保留了NKG2D结合的α1/α2结构域,与二苯并环胱氨酸(DBCO)偶联,用于叠氮修饰的肿瘤细胞的生物正交标记。同时,白细胞介素-15 (IL-15)被包裹在沸石咪唑盐框架(ZIF-8)中,以响应性释放,维持NK细胞功能并上调NKG2D受体的表达。这种协同设计为NK细胞识别和激活创造了一个正反馈回路。因此,该策略抑制了多种肿瘤类型并改善了免疫微环境。在机制上,先天免疫和适应性免疫都是通过树突状细胞和T细胞之间的协调串音来调动的,NK细胞衍生的趋化因子分泌促进了这一过程。与抗pd -1抗体联合,ZIL15-D-trMULT1诱导持久免疫记忆,抑制肿瘤远端生长和肺转移。我们的工作揭示了NK细胞通过上调不可切割的NKG2D配体密度触发的自我维持免疫回路,为对抗实体肿瘤提供了一种强大的策略。
{"title":"Bio-orthogonal truncated NKG2D ligand-based nano-igniter unleashes a self-sustaining antitumor immune circuit via NK cell activation.","authors":"Yao Gong, Xiaoying Zhang, Wenlong Ren, Jian Peng, Miaoshu Liu, Lanxin Jiang, Caixia Pei, Xingping Hu, Jie Xu, Ting Zhang, Fengxia Gao, Siqiao Li, Li Du, Wei Cheng","doi":"10.1016/j.jconrel.2026.114603","DOIUrl":"https://doi.org/10.1016/j.jconrel.2026.114603","url":null,"abstract":"<p><p>Despite the crucial role of natural killer (NK) cells in initiating antitumor immune circuits, their efficacy is limited by tumor immune evasion mechanisms, including low surface density or proteolytic shedding of activating ligands and immunosuppression. Here, we constructed a bioorthogonal truncated NKG2D (natural killer group 2, member D) ligand-based nano-igniter (ZIL15-D-trMULT1) to synchronously enhance NK cell recognition and function, thereby igniting antitumor immune circuits. This system incorporates a truncated NKG2D ligand (trMULT1) lacking the cleavable α3 domain while retaining the NKG2D-binding α1/α2 domains, conjugated to dibenzocyclooctyne (DBCO) for bioorthogonal tagging of azide-modified tumor cells. Concurrently, interleukin-15 (IL-15) was encapsulated within zeolitic imidazolate frameworks (ZIF-8) for responsive release, sustaining NK cell function and upregulating NKG2D receptor expression. This synergistic design created a positive feedback loop for NK cell recognition and activation. Consequently, this strategy suppressed multiple tumor types and improved the immune microenvironment. Mechanistically, both innate and adaptive immunity were mobilized by orchestrating crosstalk between dendritic cells and T cells, facilitated by NK cell-derived chemokine secretion. In combination with anti-PD-1 antibody, ZIL15-D-trMULT1 induced durable immune memory, restraining distal tumor growth and lung metastasis. Our work unveils a self- sustaining immune circuit triggered by NK cells through upregulating the un-cleavable NKG2D ligand density, providing a robust strategy against solid tumors.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":"114603"},"PeriodicalIF":11.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933645","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}
Disrupting mitochondrial metabolism and reactivating antitumor immunity offers a compelling strategy to enhance therapeutic outcomes in glioblastoma (GBM). Here, we report a mitochondria-targeting nanoplatform that integrates GBM-selective delivery, metabolic disruption, and immune activation for synergistic GBM therapy. The nanoplatform co-encapsulates L820-a conjugate of lonidamine (LND) and IR820 with mitochondrial affinity-and the immunomodulator TP5 into a ZIF-8 framework, which is coated with H-ferritin (HFn) for TfR1-mediated blood-brain barrier (BBB) penetration and GBM targeting. After endocytosis and GBM-specific, acid-triggered degradation, L820 accumulates in mitochondria and causes profound mitochondrial dysfunction, including membrane potential collapse, ATP depletion, and AMPK activation. Concurrently, Zn2+ released from ZIF-8 suppresses GLUT1 and HIF-1α, impairing glycolysis and reducing CD47 expression. These dual metabolic stresses induce mitochondrial DNA (mtDNA) release and activate the cGAS-STING pathway, promoting type I interferon production and immunogenic cell death. TP5 enhances T cell activation while suppressing Tregs, further remodeling the GBM immune microenvironment. This mitochondria-focused strategy achieves potent metabolic interference and immune reprogramming for effective GBM treatment.
{"title":"H-ferritin engineered nanoplatform reprograms metabolism and immunity for glioblastoma immunotherapy.","authors":"Jing Zuo, Yichun Huang, Hailong Tian, Siyuan Qin, Yonghao Yan, Han Yan, Yining Jiang, Lei Li, Shiqi Wang, Yongfeng Jia, Yuan Zhao, Canhua Huang","doi":"10.1016/j.jconrel.2026.114613","DOIUrl":"https://doi.org/10.1016/j.jconrel.2026.114613","url":null,"abstract":"<p><p>Disrupting mitochondrial metabolism and reactivating antitumor immunity offers a compelling strategy to enhance therapeutic outcomes in glioblastoma (GBM). Here, we report a mitochondria-targeting nanoplatform that integrates GBM-selective delivery, metabolic disruption, and immune activation for synergistic GBM therapy. The nanoplatform co-encapsulates L820-a conjugate of lonidamine (LND) and IR820 with mitochondrial affinity-and the immunomodulator TP5 into a ZIF-8 framework, which is coated with H-ferritin (HFn) for TfR1-mediated blood-brain barrier (BBB) penetration and GBM targeting. After endocytosis and GBM-specific, acid-triggered degradation, L820 accumulates in mitochondria and causes profound mitochondrial dysfunction, including membrane potential collapse, ATP depletion, and AMPK activation. Concurrently, Zn<sup>2+</sup> released from ZIF-8 suppresses GLUT1 and HIF-1α, impairing glycolysis and reducing CD47 expression. These dual metabolic stresses induce mitochondrial DNA (mtDNA) release and activate the cGAS-STING pathway, promoting type I interferon production and immunogenic cell death. TP5 enhances T cell activation while suppressing Tregs, further remodeling the GBM immune microenvironment. This mitochondria-focused strategy achieves potent metabolic interference and immune reprogramming for effective GBM treatment.</p>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":" ","pages":"114613"},"PeriodicalIF":11.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145933590","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}
Pub Date : 2026-01-06DOI: 10.1016/j.jconrel.2026.114607
Moritz Marschhofer , Siyu Chen , Müge Molbay , Benjamin Winkeljann , Ersilia Villano , Corinne Giancaspro , Alexandra Kourou , Otto Berninghausen , Susanne Rieder , Charlotte Ungewickell , Roland Beckmann , Bastian Popper , Ana Maria Torres , Anxo Vidal , Olivia M. Merkel , Simone P. Carneiro
KRAS G12S mutations in non-small cell lung cancer (NSCLC) remain refractory to current targeted therapies, with few clinical options and frequent resistance. While CRISPR/Cas9 enables mutation-specific gene disruption, its pulmonary application is limited by systemic clearance, hepatic tropism, and airway mucus barriers. Here, we present lipid nanoparticles (LNPs) specifically engineered for pulmonary delivery of Cas9 mRNA and KRAS G12S-targeting sgRNA, optimized through mRNA surrogate screening and orthogonal mixture design to guide lipid composition and Cas9:sgRNA weight-to-weight ratios. Two lead LNP formulations, A6 3:1 and A8 1:1, exhibited robust critical quality attributes, including particle sizes below 120 nm, low polydispersity, near-neutral zeta potential, and over 80 % encapsulation efficiency. Cryo-TEM revealed distinct morphologies correlated with enhanced transfection. In vitro, A8 1:1 achieved up to 90 % on-target gene editing in A549 cells and a 3.6-fold increase in apoptosis, while A6 3:1 induced a 3.7-fold apoptotic response. Both formulations efficiently traversed airway mucus in air-liquid interface cultures and preserved over 80 % cell viability across doses. In vivo, repeated pulmonary administration was well tolerated, with no signs of systemic toxicity or cytokine elevation in healthy or tumor-bearing mice. In an orthotopic A549-luc lung tumor model, intratracheal delivery of A6 3:1 and A8 1:1 modestly suppressed tumor growth, with histological evidence of tumor cell apoptosis for A8 1:1. Quantification confirmed a statistically significant increase of apoptosis in the A8 1:1 group, consistent with effective KRAS disruption in vivo. Overall, lead LNPs, particularly A8 1:1, enabled efficient and localized RNA-based gene editing that induced downstream apoptotic signaling, demonstrating a preliminary, yet promising, proof-of-concept for CRISPR/Cas9 therapy in NSCLC.
{"title":"Optimized lipid nanoparticles for pulmonary delivery of CRISPR/Cas9 targeting KRAS G12S in lung cancer","authors":"Moritz Marschhofer , Siyu Chen , Müge Molbay , Benjamin Winkeljann , Ersilia Villano , Corinne Giancaspro , Alexandra Kourou , Otto Berninghausen , Susanne Rieder , Charlotte Ungewickell , Roland Beckmann , Bastian Popper , Ana Maria Torres , Anxo Vidal , Olivia M. Merkel , Simone P. Carneiro","doi":"10.1016/j.jconrel.2026.114607","DOIUrl":"10.1016/j.jconrel.2026.114607","url":null,"abstract":"<div><div><em>KRAS</em> G12S mutations in non-small cell lung cancer (NSCLC) remain refractory to current targeted therapies, with few clinical options and frequent resistance. While CRISPR/Cas9 enables mutation-specific gene disruption, its pulmonary application is limited by systemic clearance, hepatic tropism, and airway mucus barriers. Here, we present lipid nanoparticles (LNPs) specifically engineered for pulmonary delivery of Cas9 mRNA and KRAS G12S-targeting sgRNA, optimized through mRNA surrogate screening and orthogonal mixture design to guide lipid composition and Cas9:sgRNA weight-to-weight ratios. Two lead LNP formulations, A6 3:1 and A8 1:1, exhibited robust critical quality attributes, including particle sizes below 120 nm, low polydispersity, near-neutral zeta potential, and over 80 % encapsulation efficiency. Cryo-TEM revealed distinct morphologies correlated with enhanced transfection. <em>In vitro</em>, A8 1:1 achieved up to 90 % on-target gene editing in A549 cells and a 3.6-fold increase in apoptosis, while A6 3:1 induced a 3.7-fold apoptotic response. Both formulations efficiently traversed airway mucus in air-liquid interface cultures and preserved over 80 % cell viability across doses. <em>In vivo</em>, repeated pulmonary administration was well tolerated, with no signs of systemic toxicity or cytokine elevation in healthy or tumor-bearing mice. In an orthotopic A549-luc lung tumor model, intratracheal delivery of A6 3:1 and A8 1:1 modestly suppressed tumor growth, with histological evidence of tumor cell apoptosis for A8 1:1. Quantification confirmed a statistically significant increase of apoptosis in the A8 1:1 group, consistent with effective <em>KRAS</em> disruption <em>in vivo</em>. Overall, lead LNPs, particularly A8 1:1, enabled efficient and localized RNA-based gene editing that induced downstream apoptotic signaling, demonstrating a preliminary, yet promising, proof-of-concept for CRISPR/Cas9 therapy in NSCLC.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"391 ","pages":"Article 114607"},"PeriodicalIF":11.5,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145902494","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}
Pub Date : 2026-01-05DOI: 10.1016/j.jconrel.2026.114601
Chang Liu , Ziwei Zhang , Ye Yuan , Hezhi Wang , Jian Guan , Shirui Mao
Immunotherapeutic failures in non-small cell lung cancer (NSCLC) and other solid tumors are often attributed to tumor-induced T cell dysfunction. However, current treatments frequently fail to initiate a complete cancer-immunity cycle within the immunosuppressive tumor microenvironment (TME). Inspired by intercellular RNA transfer mediated by extracellular vesicles (EVs), we developed an inhalable nanoplatform (p53 mRNA/lipid liquid nanoparticle complex, known as LLX) to “relay” delivery of p53 mRNA from tumor-associated macrophages (TAMs) to NSCLC cells, overcoming the challenges posed by passive diffusion and size limitations within the solid TME. After inhalation in a lung orthotopic tumor model, LLX uniformly dispersed throughout the lung airways and markedly boosted LLX-EV production by TAMs. This in vivo strategy of generating endogenous EVs loaded with exogenous mRNA not only induced p53-mediated apoptosis in NSCLC cells but also fundamentally reshaped the TME by directly modulating the TAM phenotype and alleviating T cell exhaustion. Our results show that p53 mRNA/LLX resulted in sustained tumor regression, underscoring its potential to expand the therapeutic scope of gene immunotherapy by effectively tackling key challenges in solid tumor treatment, such as immunosuppression and the limited transfection efficiency of mRNA.
{"title":"An inhalable liquid-core lipid nanoplatform enables macrophage-mediated mRNA delivery to lung tumors","authors":"Chang Liu , Ziwei Zhang , Ye Yuan , Hezhi Wang , Jian Guan , Shirui Mao","doi":"10.1016/j.jconrel.2026.114601","DOIUrl":"10.1016/j.jconrel.2026.114601","url":null,"abstract":"<div><div>Immunotherapeutic failures in non-small cell lung cancer (NSCLC) and other solid tumors are often attributed to tumor-induced T cell dysfunction. However, current treatments frequently fail to initiate a complete cancer-immunity cycle within the immunosuppressive tumor microenvironment (TME). Inspired by intercellular RNA transfer mediated by extracellular vesicles (EVs), we developed an inhalable nanoplatform (p53 mRNA/lipid liquid nanoparticle complex, known as LLX) to “relay” delivery of p53 mRNA from tumor-associated macrophages (TAMs) to NSCLC cells, overcoming the challenges posed by passive diffusion and size limitations within the solid TME. After inhalation in a lung orthotopic tumor model, LLX uniformly dispersed throughout the lung airways and markedly boosted LLX-EV production by TAMs. This <em>in vivo</em> strategy of generating endogenous EVs loaded with exogenous mRNA not only induced p53-mediated apoptosis in NSCLC cells but also fundamentally reshaped the TME by directly modulating the TAM phenotype and alleviating T cell exhaustion. Our results show that p53 mRNA/LLX resulted in sustained tumor regression, underscoring its potential to expand the therapeutic scope of gene immunotherapy by effectively tackling key challenges in solid tumor treatment, such as immunosuppression and the limited transfection efficiency of mRNA.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"391 ","pages":"Article 114601"},"PeriodicalIF":11.5,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145902495","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}
Pub Date : 2026-01-04DOI: 10.1016/j.jconrel.2025.114598
Ye Zeng , Gangyin Zhao , Shidi Wu , Bochuan Hu , Gabriel Forn-Cuní , Renzo Knol , Abdoelwaheb El Ghalbzouri , Ewa Snaar-Jagalska , Alexander Kros
Skin cancer is the third most common malignancy, with melanoma being the most challenging due to its resistance to current therapies. Gene editing technologies like CRISPR/Cas9 offer a promising strategy for targeting cancer-specific genes, but the efficient delivery of these tools to tumor sites remains a significant challenge. Lipid nanoparticles (LNPs) have emerged as the leading platform for gene editing tools due to their ability to protect and transport large payloads. To enhance the precision of gene editing in melanoma, we developed CD44-specific peptide-modified LNPs for targeted delivery of CRISPR/Cas9 mRNA and guide RNA against polo-like kinase 1 (sgPLK1). Our approach led to enhanced targeting and gene editing efficacy by specifically delivering CRISPR/Cas9 and sgPLK1 to melanoma tumor cells, resulting in significant inhibition of tumor growth in both in vitro and in vivo skin melanoma models. Moreover, this platform showed the capacity to reach metastatic melanoma in the brain and resulting in substantial suppression of tumor growth in brain metastasis models. We envision that this peptide-modification strategy could be further employed to improve the targeting capabilities and therapeutic outcomes of LNPs for CRISPR/Cas9-based gene editing, paving the way for more precise and effective cancer treatments.
{"title":"CD44-targeted lipid nanoparticles for enhanced CRISPR/Cas9 delivery in cancer gene editing","authors":"Ye Zeng , Gangyin Zhao , Shidi Wu , Bochuan Hu , Gabriel Forn-Cuní , Renzo Knol , Abdoelwaheb El Ghalbzouri , Ewa Snaar-Jagalska , Alexander Kros","doi":"10.1016/j.jconrel.2025.114598","DOIUrl":"10.1016/j.jconrel.2025.114598","url":null,"abstract":"<div><div>Skin cancer is the third most common malignancy, with melanoma being the most challenging due to its resistance to current therapies. Gene editing technologies like CRISPR/Cas9 offer a promising strategy for targeting cancer-specific genes, but the efficient delivery of these tools to tumor sites remains a significant challenge. Lipid nanoparticles (LNPs) have emerged as the leading platform for gene editing tools due to their ability to protect and transport large payloads. To enhance the precision of gene editing in melanoma, we developed CD44-specific peptide-modified LNPs for targeted delivery of CRISPR/Cas9 mRNA and guide RNA against polo-like kinase 1 (sgPLK1). Our approach led to enhanced targeting and gene editing efficacy by specifically delivering CRISPR/Cas9 and sgPLK1 to melanoma tumor cells, resulting in significant inhibition of tumor growth in both <em>in vitro</em> and <em>in vivo</em> skin melanoma models. Moreover, this platform showed the capacity to reach metastatic melanoma in the brain and resulting in substantial suppression of tumor growth in brain metastasis models. We envision that this peptide-modification strategy could be further employed to improve the targeting capabilities and therapeutic outcomes of LNPs for CRISPR/Cas9-based gene editing, paving the way for more precise and effective cancer treatments.</div></div>","PeriodicalId":15450,"journal":{"name":"Journal of Controlled Release","volume":"391 ","pages":"Article 114598"},"PeriodicalIF":11.5,"publicationDate":"2026-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145897327","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}