1
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
2
|
Wang T, Liu H, Li M, Ji Z, Zhang X, Wang N, Chen Y, Sun J, Liu F. Microneedle-based nanodrugs for tumor immunotherapy. J Control Release 2025; 380:539-562. [PMID: 39923854 DOI: 10.1016/j.jconrel.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/08/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Microneedles have emerged as a promising and effective method for delivering therapeutic drugs and immunobiologics to treat various diseases. It is widely recognized that immune therapy has limited efficacy in solid tumors due to physical barriers and the immunosuppressive tumor microenvironment. Microneedle-based nanodrugs (NDMNs) offer a novel approach to overcome these limitations. These tiny needles are designed to load a variety of inorganic and organic nanoparticles, antigen vaccines, gene drugs, oncolytic viruses, and more. Utilizing microneedle arrays, NDMNs can effectively penetrate the skin barrier, delivering drugs precisely to the tumor site or immunoactive regions within the skin. Additionally, by designing and optimizing the microneedle structure, shape, and functionality, NDMNs enable precise drug release and efficient penetration, thereby enhancing the efficacy of tumor immunotherapy. In this review, we comprehensively discuss the pivotal role of NDMNs in cancer immunotherapy, summarizing innovative microneedle design strategies, mechanisms of immune activation, and delivery strategies of various nanodrugs. Furthermore, we explore the current clinical realities, limitations, and future prospects of NDMNs in tumor immunotherapy.
Collapse
Affiliation(s)
- Tianye Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Department of General Surgery, The First Hospital of Dalian Medical University, Dalian 116000, China
| | - Hongyu Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Meng Li
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Zao Ji
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Xinyuan Zhang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Nan Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Ying Chen
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang 110001, China; Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China.
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems Ministry of Education, Shenyang 110016, China.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital, China Medical University, Shenyang 110001, China.
| |
Collapse
|
3
|
Tiwade PB, Fung V, VanKeulen-Miller R, Narasipura EA, Ma Y, Fenton OS. Non-Viral RNA Therapies for Non-Small Cell Lung Cancer and Their Corresponding Clinical Trials. Mol Pharm 2025; 22:1752-1774. [PMID: 40131145 DOI: 10.1021/acs.molpharmaceut.4c00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Ribonucleic acid (RNA)-based therapies represent a promising class of drugs for the treatment of non-small cell lung cancer (NSCLC) due to their ability to modulate gene expression. Therapies leveraging small interfering RNA (siRNA), messenger RNA (mRNA), microRNA (miRNA), and antisense oligonucleotides (ASOs) offer various advantages over conventional treatments, including the ability to target specific genetic mutations and the potential for personalized medicine approaches. However, the clinical translation of these therapeutics for the treatment of NSCLC faces challenges in delivery due to their immunogenicity, negative charge, and large size, which can be mitigated with delivery platforms. In this review, we provide a description of the pathophysiology of NSCLC and an overview of RNA-based therapeutics, specifically highlighting their potential application in the treatment of NSCLC. We discuss relevant classes of RNA and their therapeutic potential for NSCLC. We then discuss challenges in delivery and non-viral delivery strategies such as lipid- and polymer-based nanoparticles that have been developed to address these issues in preclinical models. Furthermore, we provide a summary table of clinical trials that leverage RNA therapies for NSCLC [which includes their National Clinical Trial (NCT) numbers] to highlight the current progress in NSCLC. We also discuss how these NSCLC therapies can be integrated with existing treatment modalities to enhance their efficacy and improve patient outcomes. Overall, we aim to highlight non-viral strategies that tackle RNA delivery challenges while showcasing RNA's potential as a next-generation therapy for NSCLC treatment.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/therapy
- Lung Neoplasms/drug therapy
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
- RNA, Small Interfering/administration & dosage
- Oligonucleotides, Antisense/therapeutic use
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/administration & dosage
- Clinical Trials as Topic
- Animals
- Nanoparticles/chemistry
- MicroRNAs/genetics
- MicroRNAs/therapeutic use
- RNA, Messenger/genetics
- Genetic Therapy/methods
- Drug Delivery Systems/methods
Collapse
Affiliation(s)
- Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eshan Amruth Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
4
|
Rodríguez DA, Lefebvre GPR, Yang Q, Barendrecht AD, Seinen CW, Schiffelers RM, Vader P. Incorporation of cellular membrane protein extracts into lipid nanoparticles enhances their cellular uptake and mRNA delivery efficiency. J Control Release 2025; 382:113676. [PMID: 40187649 DOI: 10.1016/j.jconrel.2025.113676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
mRNA therapeutics enable transient expression of desired proteins within cells, holding great potential for advancements in vaccines, protein replacement therapies and gene editing approaches. Lipid nanoparticles (LNPs) are arguably the leading nanoplatform for mRNA delivery due to their scalability and transfection efficiency. However, their limited ability to target specific cell types, inefficient cellular uptake by many cell types, and endosomal entrapment represent challenges for improving targeted mRNA delivery. To address this, we evaluated a novel class of LNPs functionalized with cell-derived membrane proteins, that we refer to as hybrisomes. Membrane protein extracts (MPEs) were isolated from cultured cells using a mild detergent-based extraction protocol. Cy5-labeled mRNA encoding for eGFP was used to form LNPs and hybrisomes to investigate their internalization efficiency and mRNA delivery via flow cytometry and microscopy, with MPE content incorporated into hybrisomes during microfluidic mixing. MPEs were successfully incorporated into the lipid membrane of hybrisomes. Remarkably, the cellular uptake of hybrisomes was up to 15-fold higher than LNPs, while the mRNA delivery efficiency improved up to 8-fold depending on the MPE content incorporated into the hybrisomes. Further studies confirmed that the enhanced cellular uptake of hybrisomes and mRNA is partially explained by the presence of membrane proteins and hybrisomes' unique morphology including bleb-like structures. Moreover, the versatility of hybrisomes was demonstrated by producing formulations using MPEs isolated from different cell types, which led to variations in cellular uptake and mRNA delivery, suggesting that the cell type from which MPEs are derived influences their biological function. These findings pave the way for the development of more targeted and effective nanotherapeutic strategies.
Collapse
Affiliation(s)
- Diego A Rodríguez
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gaspard P R Lefebvre
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Qiangbing Yang
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Arjan D Barendrecht
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Cor W Seinen
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Raymond M Schiffelers
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Pieter Vader
- CDL Research, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
5
|
Su J, Zhang J, Feng X, Liu J, Gao S, Liu X, Yang M, Chen Z. A universal viral capsid protein based one step RNA synthesis and packaging system for rapid and efficient mRNA vaccine development. Mol Ther 2025; 33:1720-1734. [PMID: 40022448 PMCID: PMC11997475 DOI: 10.1016/j.ymthe.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025] Open
Abstract
The success of coronavirus disease 2019 mRNA vaccines highlights the transformative potential of mRNA technology. Current mRNA vaccine development involves complex steps, including plasmid construction, RNA transcription, 5' capping, poly(A) tailing, and lipid nanoparticle encapsulation, yet challenges in vaccine accessibility persist. Here, we present an innovative mRNA platform leveraging the self-assembly capabilities of the MS2 bacteriophage viral capsid protein (VCP). A dual-promoter plasmid has been designed where one promoter drives VCP expression while the other transcribes target RNA containing pac sites, enabling rapid mRNA self-assembly in Escherichia coli. Using an ovalbumin (OVA)-based tumor model, we validate the efficacy of this system. Tumor growth is significantly inhibited, accompanied by robust immune activation. Flow cytometry analyses reveal increased frequencies of OVA-specific CD8+, as well as activated and memory T cells. Additionally, the MS2-OVA vaccine favorably modulated the tumor immunosuppressive microenvironment by reducing myeloid-derived suppressor cells, while sustained antibody responses demonstrated the platform's ability to induce durable humoral immunity. These findings establish the feasibility of one-step mRNA synthesis and packaging in E. coli, providing a versatile and rapid platform for mRNA vaccine development, with broad implications for addressing global vaccination challenges.
Collapse
Affiliation(s)
- Jiayue Su
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
| | - Jinsong Zhang
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
| | - Xiangning Feng
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
| | - Jinsong Liu
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
| | - Shan Gao
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
| | - Xinrui Liu
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
| | - Mingwei Yang
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
| | - Zeliang Chen
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China.
| |
Collapse
|
6
|
Wang J, Guo Q, He L, Song R, Du J, Zhou H, Hao Y, Yang X, Wang F, Li K, Li M, Yang Z, Sun L, Liu Z. A Nanoradiosensitizer Potentiates Tumor Radiotherapy through JFK Inhibition and Hypoxia Alleviation. NANO LETTERS 2025; 25:5435-5443. [PMID: 40125668 DOI: 10.1021/acs.nanolett.5c00677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Radiotherapy (RT) is a primary treatment for breast cancer, but its effectiveness is often compromised by hypoxia and intrinsic resistance mechanisms. The F-box protein JFK is overexpressed in breast cancer and is associated with reduced radiosensitivity, but specific JFK inhibitors are currently unavailable. Herein, we developed spherical nanoparticles (SNP-JC) designed to co-deliver small interfering RNA targeting JFK and catalase to the tumor, aiming to silence JFK and alleviate hypoxia to overcome RT resistance. Positron emission tomography imaging demonstrated that SNP-JC efficiently accumulated in the tumors. SNP-JC significantly increased DNA damage in tumor cells after RT and promoted the immunogenic cell death. The combination of SNP-JC and RT activated CD8+ T cells and elicited a robust antitumor immunity, resulting in suppressed primary tumor growth and reduced lung metastasis. Our findings demonstrate that a nanoplatform capable of simultaneously silencing JFK and mitigating hypoxia can enhance tumor radiosensitivity, improve antitumor efficacy, and prevent metastasis.
Collapse
Affiliation(s)
- Jianze Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Qianrui Guo
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Lin He
- Department of Biochemistry, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Rui Song
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Jinhong Du
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Haoyi Zhou
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Yameng Hao
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Xiujie Yang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Feng Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Kui Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Zhi Yang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Luyang Sun
- Department of Biochemistry, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Zhaofei Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, and Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing 100191, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital and Institute, Beijing 100142, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Nuclear Medicine, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
7
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
8
|
Tarannum M, Ding X, Barisa M, Hu S, Anderson J, Romee R, Zhang J. Engineering innate immune cells for cancer immunotherapy. Nat Biotechnol 2025; 43:516-533. [PMID: 40229380 DOI: 10.1038/s41587-025-02629-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 04/16/2025]
Abstract
Innate immune cells, including natural killer cells, macrophages and γδ T cells, are gaining prominence as promising candidates for cancer immunotherapy. Unlike conventional T cells, these cells possess attributes such as inherent antitumor activity, rapid immune responses, favorable safety profiles and the ability to target diverse malignancies without requiring prior antigen sensitization. In this Review, we examine the engineering strategies used to enhance their anticancer potential. We discuss challenges associated with each cell type and summarize insights from preclinical and clinical work. We propose strategies to address existing barriers, providing a perspective on the advancement of innate immune engineering as a powerful modality in anticancer treatment.
Collapse
Affiliation(s)
- Mubin Tarannum
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Xizhong Ding
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sabrina Hu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Rizwan Romee
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
| | - Jin Zhang
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
9
|
Adhikari A, Chen IA. Antibody-Nanoparticle Conjugates in Therapy: Combining the Best of Two Worlds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409635. [PMID: 40051146 PMCID: PMC12001320 DOI: 10.1002/smll.202409635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/02/2025] [Indexed: 04/17/2025]
Abstract
Monoclonal antibodies (mAbs) and antibody fragments have revolutionized medicine as highly specific binding agents and inhibitors. At the same time, several types of nanomaterials, including liposomes, lipid nanoparticles (NPs), polymersomes, metal and metal oxide NPs, and protein nanostructures, are increasingly utilized and explored for therapeutic potential due to their versatility, chemical and physical properties, and tunability. However, nanomaterials alone often lack specificity, leading to relatively low efficacy and/or high toxicity. To address this problem, a rapidly emerging area is antibody-nanomaterial conjugates (ANCs), which combine the precise targeting specificity of antibodies with the effector functionality of the nanomaterial. In this review, we give a brief introduction to mAbs and major conjugation techniques, describe major classes of nanomaterials being studied for therapeutic potential, and review the literature on ANCs of each class. Special focus is given to emerging applications including ANCs addressing the blood-brain barrier, ANCs delivering nucleic acids, and light-activated ANCs. While many disease targets are related to cancer, ANCs are also under development to address autoimmune, neurological, and infectious diseases. While important challenges remain, ANCs are poised to become a next-generation therapeutic technology.
Collapse
Affiliation(s)
- Aniruddha Adhikari
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| | - Irene A. Chen
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| |
Collapse
|
10
|
Cao L, Liu Y, Lin G. Strategies for Altering Delivery Technologies to Optimize CAR Therapy. Int J Mol Sci 2025; 26:3206. [PMID: 40244018 PMCID: PMC11989270 DOI: 10.3390/ijms26073206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been proven to be an effective strategy for the treatment of hematological malignancies. At present, how to prepare CAR-T cells efficiently, quickly, and safely is one of the urgent problems to be solved. The durability and activity of engineered T cells in solid tumors need to be further improved, and the strategy of T cells penetrating the tumor microenvironment also needs to be improved. In addition, although the problems mainly caused by T-cell biology are being solved, the manufacturing mode and process still need to be improved to ensure that CAR-T cell therapy can be widely used. This paper summarizes some strategies that can improve the efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Lili Cao
- Student Counseling Center, Shandong University, Jinan 250012, China;
| | - Yingying Liu
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| |
Collapse
|
11
|
Zheng Y, Wang B, Cai Z, Lai Z, Yu H, Wu M, Liu X, Zhang D. Tailoring nanovectors for optimal neoantigen vaccine efficacy. J Mater Chem B 2025; 13:4045-4058. [PMID: 40042164 DOI: 10.1039/d4tb02547d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
The primary objective of neoantigen vaccines is to elicit a robust anti-tumor immune response by generating neoantigen-specific T cells that can eradicate tumor cells. Despite substantial advancements in personalized neoantigen prediction using next-generation sequencing, machine learning, and mass spectrometry, challenges remain in efficiently expanding neoantigen-specific T cell populations in vivo. This challenge impedes the widespread clinical application of neoantigen vaccines. Nanovector-based neoantigen delivery systems have emerged as a promising solutions to this challenge. These nanovectors offer several advantages, such as enhanced stability, targeted intracellular delivery, sustained release, and improved antigen-presenting cell (APC) activation. Notably, they effectively deliver various neoantigen vaccine formulations (DC cell-based, synthetic long peptide (SLP)-based or DNA/mRNA-based) to APCs or T cells, thereby activating both CD4+ T and CD8+ T cells. This ultimately induces a specific anti-tumor immune response. This review focuses on recent innovations in neoantigen vaccine delivery vectors. We aim to identify optimal design parameters for vectors tailored to different neoantigen vaccine types, with an emphasis on enhancing the tumor microenvironment and stimulating the production of neoantigen-specific cytotoxic T cells. By maximizing the potential of these delivery systems, we aim to accelerate the clinical translation of neoantigen nanovaccines and advance cancer immunotherapy.
Collapse
Affiliation(s)
- Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Bing Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Zisen Lai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Haijun Yu
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| |
Collapse
|
12
|
Liu L, Kim JH, Li Z, Sun M, Northen T, Tang J, Mcintosh E, Karve S, DeRosa F. PEGylated lipid screening, composition optimization, and structure-activity relationship determination for lipid nanoparticle-mediated mRNA delivery. NANOSCALE 2025. [PMID: 40131321 DOI: 10.1039/d5nr00433k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Lipid nanoparticles (LNPs) have emerged as effective carriers for mRNA delivery in vaccine and therapeutic applications, attracting substantial attention since the COVID-19 pandemic. Continued efforts are crucial to optimize LNP composition for improved delivery efficacy and to elucidate the underlying mechanisms driving differences in protein expression. This study systematically screened PEGylated lipids for intramuscular mRNA delivery, followed by optimization of the formulation composition, physicochemical characterization, and investigation of the structure-activity relationship (SAR). Using a model ionizable lipid, we initially evaluated twenty-nine PEGylated lipids from four lipid families (glyceride, phosphoethanolamine (PE), cholesterol, and ceramide), each varying in linker chemistries, tail structures, or PEG molecular weights. 1,2-Dimyristoyl-rac-glycero-3-methoxypolyethylene glycol - 5000 (DMG-PEG5k) was identified as a promising candidate from this screening. Using a design of experiments (DoE) approach, we further optimized the formulation to increase in vivo transfection efficacy, achieving an increase in protein expression over the DMG-PEG2k benchmark. To explore the SAR of the DoE formulations, advanced physicochemical characterization was conducted including Laurdan assay, SAXS, Cryo-TEM, and QCM-D, alongside standard LNP analysis. Among the key factors examined, high mRNA encapsulation efficiency, LNP membrane integrity (especially under acidic conditions), and ordered internal structures were identified as the critical parameters for transfection efficiency. mRNA encapsulation efficiency increased with a lower PEG-lipid fraction. LNP membrane integrity, assessed by the generalized polarization (GP) ratio at pH 7.5 and 4.5 from the Laurdan assay, was strongly affected by the ionizable lipid ratio and, to a lesser extent, the cholesterol ratio. A lower GP7.5/GP4.5 ratio correlated with enhanced protein expression, primarily driven by a higher GP4.5 observed with lower ionizable lipid and higher cholesterol fractions. Overall, balancing the ratios of all LNP components is critical for maximizing LNP functionality. This study presents a systematic evaluation and characterization of LNPs with different PEG-lipid moieties, deepens SAR understanding, and provides valuable guidelines for rationally designing more effective next-generation LNPs.
Collapse
Affiliation(s)
- Lingyun Liu
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Jae-Heon Kim
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Zhongyu Li
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Mengwei Sun
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Trent Northen
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Jackie Tang
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Emma Mcintosh
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Shrirang Karve
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| | - Frank DeRosa
- mRNA Center of Excellence, Sanofi, Waltham, MA 02451, USA.
| |
Collapse
|
13
|
Pan X, Zhang YWQ, Dai C, Zhang J, Zhang M, Chen X. Applications of mRNA Delivery in Cancer Immunotherapy. Int J Nanomedicine 2025; 20:3339-3361. [PMID: 40125430 PMCID: PMC11928443 DOI: 10.2147/ijn.s500520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 03/08/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer treatment is continually advancing, with immunotherapy gaining prominence as a standard modality that has markedly improved the management of various malignancies. Despite these advancements, the efficacy of immunotherapy remains variable, with certain cancers exhibiting limited response and patient outcomes differing considerably. Thus, enhancing the effectiveness of immunotherapy is imperative. A promising avenue is mRNA delivery, employing carriers such as liposomes, peptide nanoparticles, inorganic nanoparticles, and exosomes to introduce mRNA cargos encoding tumor antigens, immune-stimulatory, or immune-modulatory molecules into the tumor immune microenvironment (TIME). This method aims to activate the immune system to target and eradicate tumor cells. In this review, we introduce the characteristics and limitations of these carriers and summarize the application and mechanisms of currently prevalent cargos in mRNA-based tumor treatment. Additionally, given the significant clinical application of immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR)-based cell therapies in solid tumors (including melanoma, non-small-cell lung cancer, head and neck squamous cell carcinoma, triple-negative breast cancer, gastric cancer) and leukemia, which have become first-line treatments, we highlight and discuss recent progress in combining mRNA delivery with ICIs, CAR-T, CAR-NK, and CAR-macrophage therapies. This combination enhances the targeting capabilities and efficacy of ICIs and CAR-cell-based therapies, while also mitigating the long-term off-target toxicities associated with conventional methods. Finally, we analyze the limitations of current mRNA delivery systems, such as nuclease-induced mRNA instability, immunogenicity risks, complex carrier production, and knowledge gaps concerning dosing and safety. Addressing these challenges is crucial for unlocking the potential of mRNA in cancer immunotherapy. Overall, exploring mRNA delivery enriches our comprehension of cancer immunotherapy and holds promise for developing personalized and effective treatment strategies, potentially enhancing the immune responses of cancer patients and extending their survival time.
Collapse
Affiliation(s)
- Xiaoyu Pan
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, People’s Republic of China
| | - Yang-Wen-Qing Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, People’s Republic of China
| | - Caixia Dai
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, People’s Republic of China
| | - Junyu Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, People’s Republic of China
| | - Minghe Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, People’s Republic of China
| | - Xi Chen
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, People’s Republic of China
| |
Collapse
|
14
|
Lan H, Zhao J, Yuan L, Li M, Pu X, Guo Y. Deep Clustering-Based Immunotherapy Prediction for Gastric Cancer mRNA Vaccine Development. Int J Mol Sci 2025; 26:2453. [PMID: 40141097 PMCID: PMC11941797 DOI: 10.3390/ijms26062453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Immunotherapy is becoming a promising strategy for treating diverse cancers. However, it benefits only a selected group of gastric cancer (GC) patients since they have highly heterogeneous immunosuppressive microenvironments. Thus, a more sophisticated immunological subclassification and characterization of GC patients is of great practical significance for mRNA vaccine therapy. This study aimed to find a new immunological subclassification for GC and further identify specific tumor antigens for mRNA vaccine development. First, deep autoencoder (AE)-based clustering was utilized to construct the immunological profile and to uncover four distinct immune subtypes of GC, labeled as Subtypes 1, 2, 3, and 4. Then, in silico prediction using machine learning methods was performed for accurate discrimination of new classifications with an average accuracy of 97.6%. Our results suggested significant clinicopathology, molecular, and immune differences across the four subtypes. Notably, Subtype 4 was characterized by poor prognosis, reduced tumor purity, and enhanced immune cell infiltration and activity; thus, tumor-specific antigens associated with Subtype 4 were identified, and a customized mRNA vaccine was developed using immunoinformatic tools. Finally, the influence of the tumor microenvironment (TME) on treatment efficacy was assessed, emphasizing that specific patients may benefit more from this therapeutic approach. Overall, our findings could help to provide new insights into improving the prognosis and immunotherapy of GC patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanzhi Guo
- College of Chemistry, Sichuan University, Chengdu 610064, China
| |
Collapse
|
15
|
Yuan Y, Li Y, Li G, Lei L, Huang X, Li M, Yao Y. Intelligent Design of Lipid Nanoparticles for Enhanced Gene Therapeutics. Mol Pharm 2025; 22:1142-1159. [PMID: 39878334 DOI: 10.1021/acs.molpharmaceut.4c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Lipid nanoparticles (LNPs) are an effective delivery system for gene therapeutics. By optimizing their formulation, the physiochemical properties of LNPs can be tailored to improve tissue penetration, cellular uptake, and precise targeting. The application of these targeted delivery strategies within the LNP framework ensures efficient delivery of therapeutic agents to specific organs or cell types, thereby maximizing therapeutic efficacy. In the realm of genome editing, LNPs have emerged as a potent vehicle for delivering CRISPR/Cas components, offering significant advantages such as high in vivo efficacy. The incorporation of machine learning into the optimization of LNP platforms for gene therapeutics represents a significant advancement, harnessing its predictive capabilities to substantially accelerate the research and development process. This review highlights the dynamic evolution of LNP technology, which is expected to drive transformative progress in the field of gene therapy.
Collapse
Affiliation(s)
- Yichen Yuan
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Ying Li
- Research Center for Space Computing System, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Guo Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
| | - Liqun Lei
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Xingxu Huang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yuan Yao
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
- Zhejiang Key Laboratory of Intelligent Manufacturing for Functional Chemicals, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| |
Collapse
|
16
|
Xiao X, Yang S, Jiang G, He S. Current views and trends of nanomaterials as vectors for gene delivery since the 21st century: a bibliometric analysis. Nanomedicine (Lond) 2025; 20:439-454. [PMID: 39878523 PMCID: PMC11875476 DOI: 10.1080/17435889.2025.2457781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Gene therapy is garnering increasing support due to its potential for a "once-delivered, lifelong benefit." The limitations of traditional gene delivery methods have spurred the advancement of bionanomaterials. Despite this progress, a thorough analysis of the evolution, current state, key contributors, focal studies, and future directions of nanomaterials in gene delivery remains absent. METHODS This study scrutinizes articles from the Web of Science, spanning 1 January 2 000, to 31 December 2023, employing various online tools for analysis and visualization. RESULTS The 21st century has witnessed consistent growth in scholarly work in this domain globally, with notable contributions from China and the US. At the same time, the Chinese Academy of Sciences (CAS), Harvard University, and Massachusetts Institute of Technology (MIT) have emerged as the most productive institutions, with CAS's academician Weihong Tan becoming the field's leading author. While drug delivery and nanoparticles (NPs) have been central themes for two decades, the research focus has shifted from modifying NPs and ultrafine particles to exploring polymer-hybrid NPs, mRNA vaccines, immune responses, green synthesis, and CRISPR/Cas tools. CONCLUSIONS This shift marks the transition from nanomaterials to bionanomaterials. The insights provided by this research offer a comprehensive overview of the field and valuable guidance for future investigations.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sheng Yang
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ge Jiang
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shisheng He
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
He W, Zhang M, Zhong Y, Gao Y, Fan D, Lu X. Diverse nanoparticles deliver mRNA to enhance tumor immunotherapy. BMB Rep 2025; 58:124-132. [PMID: 40058873 PMCID: PMC11955730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/15/2024] [Accepted: 01/15/2025] [Indexed: 04/01/2025] Open
Abstract
Limited efficacy and severe side effects often result in suboptimal outcomes to solid tumor therapies. In contrast, the reduced side effects and potential long-term benefits of tumor immunotherapy offer promise, notwithstanding the challenges of variable patient responses and immune-related adverse events hindering its widespread application. Recent advances in mRNA technology have revolutionized cancer immunotherapy. The versatility of mRNA as a vaccine and therapeutic agent is evident in it overcoming the limitations of traditional approaches by reducing in vivo toxicity and enhancing immune response activation. The synergy between mRNA technology and immunotherapy is increasingly being utilized to improve cancer treatment efficacy. One critical aspect of maximizing the therapeutic impact of mRNA-based treatments is the selection of an effective delivery system. Due to their size properties and material characteristics, nanoparticles offer a transformative solution, enabling the targeted and efficient delivery of mRNA to tumor tissues or immune cells. This precision delivery mechanism significantly enhances the effectiveness of immunotherapy, and represents a significant advance in cancer treatment. This review aims to explore how mRNA delivery via nanoparticles enhances tumor immunotherapy. Examination of its applications and challenges provides insights and strategic perspectives to advance this innovative therapeutic approach. [BMB Reports 2025; 58(3): 124-132].
Collapse
Affiliation(s)
- Wei He
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Meng Zhang
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Yuexia Zhong
- Outpatient Department of the Second Affiliated Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Dong Fan
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China
| | - Xiyan Lu
- Outpatient Department of the Second Affiliated Hospital, Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
18
|
Zhang M, Wang Y, Li B, Yang B, Zhao M, Li B, Liu J, Hu Y, Wu Z, Ong Y, Han X, Ding L, Zhu K, Li J, Luo M, Chen S, Peng L, Zhang L, Chen X, Ni Q. STING-Activating Polymers Boost Lymphatic Delivery of mRNA Vaccine to Potentiate Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412654. [PMID: 39713955 DOI: 10.1002/adma.202412654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/02/2024] [Indexed: 12/24/2024]
Abstract
The unprecedented success of mRNA vaccines against COVID-19 has inspired scientists to develop mRNA vaccines for cancer immunotherapy. However, using nucleoside modified mRNA as vaccine, though evading innate immune toxicity, diminishes its therapeutic efficacy for cancers. Here, we report a polyvalent stimulator of interferon genes (STING) activating polymer (termed as PD) to bolster the immunogenicity of mRNA vaccine. PD is made of tertiary amine units and conjugated with a biodegradable alkyl chain. Co-formulation of PDs bearing different number of tertiary amines with lipid materials and mRNA resulted in the lipid-like nanoparticles (PD LNPs) which effectively promoted lymphatic delivery and elicited robust immune activation via the STING signaling pathway. Notably, PD with eighteen tertiary amines (PD18) is predominant in balancing immune activity and tolerability. Subcutaneous administration of PD18 LNPs containing ovalbumin (OVA) mRNA enhanced the frequency of antigen specific CD8+ T cell with immune memory, leading to potent anticancer efficacy that surpassed 2'3'-cGAMP in both prophylactic and therapeutic cancer models. Additionally, PD18 LNP-based mRNA vaccine showed conferred resistance to cancer challenge for up to 60 days. Overall, this study offers a new perspective of using STING- activating polymer for imparting synergistic activity in mRNA vaccine-based cancer immunotherapy.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yongling Wang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, P. R. China
| | - Benhao Li
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Bowei Yang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Mengyao Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Bingyu Li
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jianping Liu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yaxin Hu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Zhaoming Wu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yenhui Ong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaolin Han
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lingwen Ding
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Kongfu Zhu
- Department of Biological Science, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117558, Singapore
| | - Jianwei Li
- Department of Biological Science, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117558, Singapore
| | - Min Luo
- Department of Biological Science, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117558, Singapore
| | - Shengqi Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Ling Peng
- Centre Interdisciplinaire de Nanoscience de Marseille, Aix-Marseille Universite, CNRS, UMR 7325, ́ "Equipe Labellisee Ligue ́Contre le Cancer", Marseille, 13288, France
| | - Longjiang Zhang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering, and Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore, 117544, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 138667, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
19
|
Li Y, Xu Y, Su W, Xu J, Ye Z, Wang Z, Liu Q, Chen F. Exploring the immuno-nano nexus: A paradigm shift in tumor vaccines. Biomed Pharmacother 2025; 184:117897. [PMID: 39921945 DOI: 10.1016/j.biopha.2025.117897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Tumor vaccines have become a crucial strategy in cancer immunotherapy. Challenges of traditional tumor vaccines include inadequate immune activation and low efficacy of antigen delivery. Nanoparticles, with their tunable properties and versatile functionalities, have redefined the landscape of tumor vaccine design. In this review, we outline the multifaceted roles of nanoparticles in tumor vaccines, ranging from their capacity as delivery vehicles to their function as immunomodulatory adjuvants capable of stimulating anti-tumor immunity. We discuss how this innovative approach significantly boosts antigen presentation by leveraging tailored nanoparticles that facilitate efficient uptake by antigen-presenting cells. These nanoparticles have been meticulously designed to overcome biological barriers, ensuring optimal delivery to lymph nodes and effective interaction with the immune system. Overall, this review highlights the transformative power of nanotechnology in redefining the principles of tumor vaccines. The intent is to inform more efficacious and precise cancer immunotherapies. The integration of these advanced nanotechnological strategies should unlock new frontiers in tumor vaccine development, enhancing their potential to elicit robust and durable anti-tumor immunity.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yike Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenwen Su
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jia Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zifei Ye
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhuoyi Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Qihui Liu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Fangfang Chen
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
20
|
Fleite S, Cassanello M, Buera MDP. Modifications of biological membranes, fat globules and liposomes promoted by cavitation processes. Consequences and applications. Chem Phys Lipids 2025; 267:105462. [PMID: 39622431 DOI: 10.1016/j.chemphyslip.2024.105462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Cavitation-based technologies, such as ultrasound (or acoustic cavitation, AC) and hydrodynamic cavitation (HC), are gaining interest among green processing technologies due to their cost effectiveness in operation, toxic solvent use reduction, and ability to obtain superior processed products, compared to conventional methods. Both AC and HC generate bubbles, but their effects may differ and it is difficult to make comparisons as both are based on different phenomena and are subject to different operational variables. AC is one of the most used techniques in extraction and homogenization processes at the laboratory level. However, upscaling to an industrial level is hard. On the other hand, HC is based on the passage of the liquid through a constriction (orifice plate, Venturi, throttling valve), which causes an increase in liquid velocity at the expense of local pressure, forcing the pressure around the contraction below the threshold pressure that induces the formation of cavities. Some applications of cavitation technologies, such as the production of liposomes or lipid nanoparticles (LNPs) allow the generation of delivery systems for biomedical applications.Many others (inactivation of pathogenic viruses, bacteria and algae for water purification, extraction procedures, third generation of biofuel production, green extractions) are based on the disruption of lipid membranes. There are also applications aimed at the modification of membranes (like the milk fat globule) for the development of innovative products. Process parameters, such as cavitation intensity, duration and temperature define the impact of the process on the physical, chemical, and biological characteristics of the membranes. Thus, the adequate implementation of cavitation processes requires understanding of interactions and synergistic mechanisms in complex systems and of their effects on membranes at the microscopic or molecular level. In the present work, the use of cavitation technologies for the generation of LNPs or nanostructured lipid carriers, and the effects of AC and HC treatments on several types of membrane systems (liposomes, solid lipid nanoparticles, milk fat globules, algae and bacterial membranes) are discussed, focusing on the structural and chemical modifications of lipidic structures under cavitation.
Collapse
Affiliation(s)
- Santiago Fleite
- CONICET - Universidad de Buenos Aires, Instituto de Tecnología de Alimentos y Procesos Químicos (ITAPROQ), Ciudad Universitaria, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Industrias, Ciudad Universitaria, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina; Universidad de Buenos Aires, Facultad de Agronomía, Cátedra de Química Inorgánica y Analítica, Argentina
| | - Miryan Cassanello
- CONICET - Universidad de Buenos Aires, Instituto de Tecnología de Alimentos y Procesos Químicos (ITAPROQ), Ciudad Universitaria, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Industrias, Ciudad Universitaria, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina
| | - María Del Pilar Buera
- CONICET - Universidad de Buenos Aires, Instituto de Tecnología de Alimentos y Procesos Químicos (ITAPROQ), Ciudad Universitaria, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Industrias, Ciudad Universitaria, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Ciudad Universitaria, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina.
| |
Collapse
|
21
|
Dong S, Li X, Huang Q, Li Y, Li J, Zhu X, Xue C, Chen R, Zeng Y, Wu J, Zhong Y, Hu S. Resistance to immunotherapy in non-small cell lung cancer: Unraveling causes, developing effective strategies, and exploring potential breakthroughs. Drug Resist Updat 2025; 81:101215. [PMID: 40081220 DOI: 10.1016/j.drup.2025.101215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Over the last two decades, advancements in deciphering the intricate interactions between oncology and immunity have fueled a meteoric rise in immunotherapy for non-small cell lung cancer, typified by an explosive growth of immune checkpoint inhibitors. However, resistance to immunotherapy remains inevitable. Herein we unravel the labyrinthine mechanisms of resistance to immunotherapy, characterized by their involvement of nearly all types of cells within the body, beyond the extrinsic cancer cells, and importantly, such cells are not only (inhibitory or excitatory, or both) signal recipients but also producers, acting in a context-dependent manner. At the molecular level, these mechanisms underlie genetic and epigenetic aberrations, which are regulated by or regulate various protein kinases, growth factors, and cytokines with inherently dynamic and spatially heterogeneous properties. Additionally, macroscopic factors such as nutrition, comorbidities, and the microbiome within and around organs or tumor cells are involved. Therefore, developing therapeutic strategies combined with distinct action informed by preclinical, clinical, and real-world evidence, such as radiotherapy, chemotherapy, targeted therapy, antibody-drug conjugates, oncolytic viruses, and cell-based therapies, may stand as a judicious reality, although the ideality is to overcome resistance point-by-point through a novel drug. Notably, we highlight a realignment of treatment aims, moving the primary focus from eliminating cancer cells -- such as through chemotherapy and radiotherapy -- to promoting immune modulation and underscore the value of regulating various components within the host macro- or micro-environment, as their effects, even if seemingly minimal, can cumulatively contribute to visible clinical benefit when applied in combination with ICIs. Lastly, this review also emphasizes the current hurdles scattered throughout preclinical and clinical studies, and explores evolving directions in the landscape of immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Xiaoyu Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Qing Huang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuanxiang Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | | | - Xianmin Zhu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Chang Xue
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Runzhi Chen
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuan Zeng
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Jingyi Wu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yi Zhong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| |
Collapse
|
22
|
Wang J, Cai L, Li N, Luo Z, Ren H, Zhang B, Zhao Y. Developing mRNA Nanomedicines with Advanced Targeting Functions. NANO-MICRO LETTERS 2025; 17:155. [PMID: 39979495 PMCID: PMC11842722 DOI: 10.1007/s40820-025-01665-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025]
Abstract
The emerging messenger RNA (mRNA) nanomedicines have sprung up for disease treatment. Developing targeted mRNA nanomedicines has become a thrilling research hotspot in recent years, as they can be precisely delivered to specific organs or tissues to enhance efficiency and avoid side effects. Herein, we give a comprehensive review on the latest research progress of mRNA nanomedicines with targeting functions. mRNA and its carriers are first described in detail. Then, mechanisms of passive targeting, endogenous targeting, and active targeting are outlined, with a focus on various biological barriers that mRNA may encounter during in vivo delivery. Next, emphasis is placed on summarizing mRNA-based organ-targeting strategies. Lastly, the advantages and challenges of mRNA nanomedicines in clinical translation are mentioned. This review is expected to inspire researchers in this field and drive further development of mRNA targeting technology.
Collapse
Affiliation(s)
- Ji Wang
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Lijun Cai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Ning Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Zhiqiang Luo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Haozhen Ren
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
| | - Yuanjin Zhao
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China.
| |
Collapse
|
23
|
Gao M, Zhong J, Liu X, Zhao Y, Zhu D, Shi X, Xu X, Zhou Q, Xuan W, Zhang Y, Zhou Y, Cheng J. Deciphering the Role of PEGylation on the Lipid Nanoparticle-Mediated mRNA Delivery to the Liver. ACS NANO 2025; 19:5966-5978. [PMID: 39899798 DOI: 10.1021/acsnano.4c09399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Organ- and cell-specific delivery of mRNA via modular lipid nanoparticles (LNPs) is promising in treating various diseases, but targeted cargo delivery is still very challenging. Most previous work focuses on screening ionizable and helper lipids to address the above issues. Here, we report the multifacial role of PEGylated lipids in manipulating LNP-mediated delivery of mRNA to the liver. We employed the typical excipients in LNP products, including DLin-MC3-DMA, DPSC, and cholesterol. Five types of PEGylated lipids were selected, and their molar ratio was fixed at 1.5% with a constant PEG molecular weight of 2000 Da. The architecture of steric lipids dramatically affected the in vitro gene transfection, in vivo blood clearance, liver deposition, and targeting of specific cells, all of which were closely linked to the de-PEGylation rate. The fast de-PEGylation resulted in short blood circulation and high accumulation in the liver. However, the ultrafast de-PEGylation enabled the deposition of more LNPs in Kupffer cells other than hepatocytes. Surprisingly, simply changing the terminal groups of PEGylated lipids from methoxyl to carboxyl or amine could dramatically increase the liver delivery of LNPs, which might be associated with the accelerated de-PEGylation rate and enhanced LNP-cell interaction. The current work highlights the importance of manipulating steric lipids in promoting mRNA delivery, offering an alternative approach for formulating and optimizing mRNA LNPs.
Collapse
Affiliation(s)
- Menghua Gao
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Jiafeng Zhong
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
| | - Xinxin Liu
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
| | - Yanjun Zhao
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Dingcheng Zhu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, Zhejiang, China
| | - Xiaohuo Shi
- Instrumentation and Service Center for Molecular Sciences, Westlake University, Hangzhou 310030, China
| | - Xuehan Xu
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
| | - Qin Zhou
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Wenjing Xuan
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yue Zhang
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
| | - Yaofeng Zhou
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Jianjun Cheng
- School of Engineering, Westlake University, Hangzhou 310030, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou 310030, Zhejiang, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| |
Collapse
|
24
|
Park W, Choi J, Hwang J, Kim S, Kim Y, Shim MK, Park W, Yu S, Jung S, Yang Y, Kweon DH. Apolipoprotein Fusion Enables Spontaneous Functionalization of mRNA Lipid Nanoparticles with Antibody for Targeted Cancer Therapy. ACS NANO 2025; 19:6412-6425. [PMID: 39908463 PMCID: PMC11841042 DOI: 10.1021/acsnano.4c16562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
The mRNA-lipid nanoparticles (mRNA@LNPs) offer a novel opportunity to treat targets previously considered undruggable. Although antibody conjugation is crucial for enhancing the specificity, delivery efficiency, and minimizing the toxicity of mRNA therapeutics, current chemical conjugation methods are complex and produce heterogeneous particles with misoriented antibodies. In this work, we introduce a chemical-free approach to functionalize mRNA@LNPs with antibodies, mimicking protein corona formation for targeted mRNA delivery. By fusing apolipoprotein to the Fc domain of a targeting antibody, we enabled the antibody to spontaneously display on the surface of mRNA@LNPs without altering the existing LNP process or employing complex chemical conjugation techniques. We demonstrated precise protein expression using trastuzumab-bound mRNA@LNPs, facilitating specific mRNA expression in HER2-positive cancer cells. mRNA was efficiently delivered to the tumor site after intravenous administration. While the control LNPs lacking targeting antibodies caused acute liver toxicity, trastuzumab-displayed LNPs showed no systemic toxicity. The tumor-specific delivery of p53 tumor suppressor mRNA led to the complete regression of cancer cells. Thus, apolipoprotein fusion enables a straightforward and scalable production of antibody-functionalized mRNA@LNPs, offering significant therapeutic potential in gene therapy.
Collapse
Affiliation(s)
- Wonbeom Park
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Jiwoong Choi
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
| | - Jaehyeon Hwang
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Suhyun Kim
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Yelee Kim
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
- Department
of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Man Kyu Shim
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
| | - Wooram Park
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Seokhyeon Yu
- Research
Center, MVRIX, Anyang 14058, Republic of Korea
| | - Sangwon Jung
- Research
Center, MVRIX, Anyang 14058, Republic of Korea
| | - Yoosoo Yang
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
- Division
of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Dae-Hyuk Kweon
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| |
Collapse
|
25
|
Somu Naidu G, Rampado R, Sharma P, Ezra A, Kundoor GR, Breier D, Peer D. Ionizable Lipids with Optimized Linkers Enable Lung-Specific, Lipid Nanoparticle-Mediated mRNA Delivery for Treatment of Metastatic Lung Tumors. ACS NANO 2025; 19:6571-6587. [PMID: 39912611 PMCID: PMC11841047 DOI: 10.1021/acsnano.4c18636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
Lipid nanoparticles (LNPs) have emerged as a groundbreaking delivery system for vaccines and therapeutic mRNAs. Ionizable lipids are the most pivotal component of LNPs due to their ability to electrostatically interact with mRNA, allowing its encapsulation while concurrently enabling its endosomal escape following cellular internalization. Thus, extensive research has been performed to optimize the ionizable lipid structure and to develop formulations that are well tolerated and allow efficient targeting of different organs that result in a high and sustained mRNA expression. However, one facet of the ionizable lipids' structure has been mostly overlooked: the linker segment between the ionizable headgroup and their tails. Here, we screened a rationally designed library of ionizable lipids with different biodegradable linkers. We extensively characterized LNPs formulated using these ionizable lipids and elucidated how these minor structural changes in the ionizable lipids structure radically influenced the LNPs' biodistribution in vivo. We showed how the use of amide and urea linkers can modulate the LNPs' pKa, resulting in an improved specificity for lung transfection. Finally, we demonstrated how one of these lipids (lipid 35) that form LNPs entrapping a bacterial toxin [pseudomonas exotoxin A (mmPE)] in the form of an mRNA reduced tumor burden and significantly increased the survival of mice with lung metastasis.
Collapse
Affiliation(s)
- Gonna Somu Naidu
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Riccardo Rampado
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Pharmaceutical Sciences, University of
Padova, Padova 35131, Italy
| | - Preeti Sharma
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Assaf Ezra
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Govinda Reddy Kundoor
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Dor Breier
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Dan Peer
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| |
Collapse
|
26
|
Kavanagh EW, Tzeng SY, Sharma N, Cutting GR, Green JJ. Ligand-free biodegradable poly(beta-amino ester) nanoparticles for targeted systemic delivery of mRNA to the lungs. Biomaterials 2025; 313:122753. [PMID: 39217793 PMCID: PMC11571037 DOI: 10.1016/j.biomaterials.2024.122753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
Non-viral nanoparticles (NPs) have seen heightened interest as a delivery method for a variety of clinically relevant nucleic acid cargoes in recent years. While much of the focus has been on lipid NPs, non-lipid NPs, including polymeric NPs, have the possibility of improved efficacy, safety, and targeting, especially to non-liver organs following systemic administration. A safe and effective systemic approach for intracellular delivery to the lungs could overcome limitations to intratracheal/intranasal delivery of NPs and improve clinical benefit for a range of diseases including cystic fibrosis. Here, engineered biodegradable poly (beta-amino ester) (PBAE) NPs are shown to facilitate efficient delivery of mRNA to primary human airway epithelial cells from both healthy donors and individuals with cystic fibrosis. Optimized NP formulations made with differentially endcapped PBAEs and systemically administered in vivo lead to high expression of mRNA within the lungs in BALB/c and C57 B/L mice without requiring a complex targeting ligand. High levels of mRNA-based gene editing were achieved in an Ai9 mouse model across bronchial, epithelial, and endothelial cell populations. No toxicity was observed either acutely or over time, including after multiple systemic administrations of the NPs. The non-lipid biodegradable PBAE NPs demonstrate high levels of transfection in both primary human airway epithelial cells and in vivo editing of lung cell types that are targets for numerous life-limiting diseases particularly single gene disorders such as cystic fibrosis and surfactant deficiencies.
Collapse
Affiliation(s)
- Erin W Kavanagh
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Neeraj Sharma
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Garry R Cutting
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Departments of Chemical & Biomolecular Engineering, Materials Science & Engineering, Neurosurgery, Oncology, and Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
27
|
Masarwy R, Breier D, Stotsky‐Oterin L, Ad‐El N, Qassem S, Naidu GS, Aitha A, Ezra A, Goldsmith M, Hazan‐Halevy I, Peer D. Targeted CRISPR/Cas9 Lipid Nanoparticles Elicits Therapeutic Genome Editing in Head and Neck Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411032. [PMID: 39736115 PMCID: PMC11831472 DOI: 10.1002/advs.202411032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/27/2024] [Indexed: 01/01/2025]
Abstract
Squamous cell carcinomas of the head and neck (HNSCC) originate in the upper aerodigestive tract, including the oral cavity, pharynx, and larynx. Current treatments of locally advanced HNSCC often lead to high treatment failure, and disease recurrence, resulting in poor survival rates. Advances in mRNA technologies and lipid nanoparticle (LNP) delivery systems led to several clinical trials involving LNP-CRISPR-Cas9 mRNA-based therapeutics. Despite these advances, achieving cell-type-specific extrahepatic mRNA delivery is still challenging. This study introduces a safe and effective intratumoral EGFR-targeted CRISPR-LNP delivery strategy for knocking out SOX2, which is a cancer-specific gene. To assess their therapeutic potential, it is shown that LNPs made from ionizable lipids with helper lipids co-encapsulating Cas9 mRNA and sgRNA targeting SOX2 (sgSOX2), lead to a ≈60% reduction in HNSCC cell viability in vitro. Next, using a xenograft HNSCC mouse model, targeted delivery of 𝜶EGFR- CRISPR-sgSOX2-LNPs to HNSCC cells resulted in a 90% inhibition of tumor growth and a 90% increase in survival for > 84 days, with tumor disappearance observed in 50% of the mice. These findings emphasize the potential of targeted mRNA-Cas9-LNPs in clinically accessible solid tumors, specifically in reaching tumor cells and inducing persistent therapeutic responses in tumors with high-recurrence rates like HNSCC.
Collapse
|
28
|
Chulanova Y, Breier D, Peer D. Delivery of genetic medicines for muscular dystrophies. Cell Rep Med 2025; 6:101885. [PMID: 39765231 PMCID: PMC11866442 DOI: 10.1016/j.xcrm.2024.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/29/2024] [Accepted: 12/05/2024] [Indexed: 01/24/2025]
Abstract
Muscular dystrophies are a group of heterogenic disorders characterized by progressive muscle weakness, the most common of them being Duchenne muscular dystrophy (DMD). Muscular dystrophies are caused by mutations in over 50 distinct genes, and many of them are caused by different genetic mechanisms. Currently, none of these diseases have a cure. However, in recent years, significant progress has been made to correct the underlying genetic cause. The clinical development of adeno-associated viral vector-based therapies has simultaneously produced excitement and disappointment in the research community due to the moderate effect, making it clear that new methods of muscle delivery have to be created. Herein, we review the main characteristics of major muscular dystrophies and outline various muscle-targeted delivery methods being explored for genetic medicines.
Collapse
Affiliation(s)
- Yulia Chulanova
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dor Breier
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
29
|
Shi G, Xu Y, Qiu H, Cao F, Xiao ZX, Zhang C, Zha GF. Personalized membrane protein vaccine based on a lipid nanoparticle delivery system prevents postoperative recurrence in colorectal cancer models. Acta Biomater 2025; 192:315-327. [PMID: 39637957 DOI: 10.1016/j.actbio.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
While accessing tumor neoantigens and developing effective delivery systems have posed significant challenges in therapeutic oncology vaccines, this study introduces a cost- and time-efficient personalized tumor vaccine demonstrating potent anti-tumor effects in a mouse xenograft model. This vaccine utilizes a lipid nanoparticle (C5 LNP) system loaded with membrane protein antigens (mAg) derived from surgically excised tumor tissue. Its safety and efficacy were validated in a B16-OVA murine model. C5/OVA exhibited significant uptake by dendritic cells (DCs), leading to cross-presentation, maturation, and subsequent initiation of robust cell-mediated and humoral immune responses. This resulted in clear tumor growth suppression and extended survival in the B16-OVA model. Furthermore, the personalized C5/mAg vaccine effectively inhibited tumor growth in a colorectal carcinoma model. When combined with anti-PD-1 therapy, it notably increased complete remission (CR) rates in the CT26 xenograft model. Vaccinated mice demonstrated 100 % resistance to tumor rechallenge, underscoring the vaccine's ability to induce long-term immune memory. This study presents a promising personalized cancer vaccine delivery system with potential for both treatment and prevention of carcinoma in clinical applications. STATEMENT OF SIGNIFICANCE: In this paper, we present a scheme for manufacturing personalized tumor vaccines. The vaccine component consists of lipid nanoparticles (LNPs) designated C5 and membrane protein antigens (mAg) derived from autologous tumor tissue surgically resected from the patient. Our study demonstrates that the personalized mAg-C5 vaccine for colorectal carcinoma significantly inhibits tumor growth. Furthermore, conjugation with anti-PD-1 therapy demonstrably increased the complete remission (CR) rate in the murine CT26 xenograft tumor model. Additionally, mice treated with the C5/mAg vaccine exhibited 100 % resistance to tumor growth in a colon carcinoma rechallenge model, indicating the induction of immune memory by the vaccine. Our research results suggest that the mAg-LNP vaccine is a simple, cost-effective treatment that clinicians can easily and quickly integrate into routine practice.
Collapse
Affiliation(s)
- Guangzhao Shi
- Department: Digestive Medicine Centre, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Yuandong Xu
- Department: Scientific Research Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Haowei Qiu
- Department: Scientific Research Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Fei Cao
- Department: Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Ze-Xiu Xiao
- Department: Research and development center, Institution: Shenzhen MagicRNA Biotech, No.459, Qiaokai Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Changhua Zhang
- Department: Digestive Medicine Centre, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China.
| | - Gao-Feng Zha
- Department: Digestive Medicine Centre, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China; Department: Scientific Research Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China.
| |
Collapse
|
30
|
Gao Z, Jing B, Wang Y, Wan W, Dong X, Liu Y. Exploring the impact of lipid nanoparticles on protein stability and cellular proteostasis. J Colloid Interface Sci 2025; 678:656-665. [PMID: 39216393 DOI: 10.1016/j.jcis.2024.08.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Lipid nanoparticles (LNPs) have become pivotal in advancing modern medicine, from mRNA-based vaccines to gene editing with CRISPR-Cas9 systems. Though LNPs based therapeutics offer promising drug delivery with satisfactory clinical safety profiles, concerns are raised regarding their potential nanotoxicity. Here, we explore the impacts of LNPs on protein stability in buffer and cellular protein homeostasis (proteostasis) in HepG2 cells. First, we show that LNPs of different polyethylene glycol (PEG) molar ratios to total lipid ratio boost protein aggregation propensity by reducing protein stability in cell lysate and blood plasma. Second, in HepG2 liver cells, these LNPs induce global proteome aggregation, as imaged by a cellular protein aggregation fluorescent dye (AggStain). Such LNPs induced proteome aggregation is accompanied by decrease in cellular micro-environmental polarity as quantified by a solvatochromic protein aggregation sensor (AggRetina). The observed local polarity fluctuations may be caused by the hydrophobic contents of LNPs that promote cellular proteome aggregation. Finally, we exploit RNA sequencing analysis (RNA-Seq) to reveal activation of unfolded protein response (UPR) pathway and other proteostasis genes upon LNPs treatment. Together, these findings highlight that LNPs may induce subtle proteome stress by compromising protein stability and proteostasis even without obvious damage to cell viability.
Collapse
Affiliation(s)
- Zifan Gao
- Dalian Medical University, Dalian 116044, China
| | - Biao Jing
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yuhui Wang
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Wang Wan
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University, Dalian 116023, China.
| | - Yu Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
31
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Cancer vaccines: platforms and current progress. MOLECULAR BIOMEDICINE 2025; 6:3. [PMID: 39789208 PMCID: PMC11717780 DOI: 10.1186/s43556-024-00241-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Cancer vaccines, crucial in the immunotherapeutic landscape, are bifurcated into preventive and therapeutic types, both integral to combating oncogenesis. Preventive cancer vaccines, like those against HPV and HBV, reduce the incidence of virus-associated cancers, while therapeutic cancer vaccines aim to activate dendritic cells and cytotoxic T lymphocytes for durable anti-tumor immunity. Recent advancements in vaccine platforms, such as synthetic peptides, mRNA, DNA, cellular, and nano-vaccines, have enhanced antigen presentation and immune activation. Despite the US Food and Drug Administration approval for several vaccines, the full therapeutic potential remains unrealized due to challenges such as antigen selection, tumor-mediated immunosuppression, and optimization of delivery systems. This review provides a comprehensive analysis of the aims and implications of preventive and therapeutic cancer vaccine, the innovative discovery of neoantigens enhancing vaccine specificity, and the latest strides in vaccine delivery platforms. It also critically evaluates the role of adjuvants in enhancing immunogenicity and mitigating the immunosuppressive tumor microenvironment. The review further examines the synergistic potential of combining cancer vaccines with other therapies, such as chemotherapy, radiotherapy, and immune checkpoint inhibitors, to improve therapeutic outcomes. Overcoming barriers such as effective antigen identification, immunosuppressive microenvironments, and adverse effects is critical for advancing vaccine development. By addressing these challenges, cancer vaccines can offer significant improvements in patient outcomes and broaden the scope of personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, 641100, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
32
|
Jin M, Wu H, Jin W, Zeng B, Liu Y, Wang N, Wang S, Chen L, Gao Z, Huang W. Transferrin Protein Corona-Targeted Codelivery of Tirapazamine and IR820 Facilitates Efficient PDT-Induced Hypoxic Chemotherapy on 4T1 Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:1892-1910. [PMID: 39699197 DOI: 10.1021/acsami.4c15045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Protein corona (PC) formation confers novel biological properties to the original nanomaterial, impeding its uptake and targeting efficacy in cells and tissues. Although many studies discussing PC formation have focused on inert proteins that may inhibit the function of nanomaterials, some functional plasma proteins with intrinsic targeting capabilities can also be adsorbed to the surface of nanomaterials, with active ligand properties to improve the targeting ability. In this approach, nanomaterials are surface-engineered to promote the adsorption of specific functional plasma proteins that are directly targeted to transport nanomaterials to the target site. In this study, T10 peptide-modified liposomes were employed to construct an in situ transferrin (Tf) PC-mediated liposome carrying a hypoxia-sensitive chemotherapy drug (tirapazamine, TPZ) and a photosensitizer (indocyanine green, IR820). The water-soluble drug TPZ was encapsulated in mesoporous silica nanoparticles (MSNs) and coated with IR820 (IR)-loaded liposome. Lipid-coated MSNs can inhibit aggregation in the body and significantly reduce the rapid release of water-soluble drugs, resulting in improved system stability and sustained release. Upon entering the in vivo circulation, T10 bound specifically to Tf in plasma to form an in situ Tf liposome-PC complex with enhanced targeting efficacy compared to traditional ligand-modified active-targeting strategies. However, large-sized PC particles faced challenges in penetrating deep into tumor tissues. IR could kill tumors through photodynamic therapy (PDT) and elicit complementary antitumor effects with the hypoxia-sensitive drug TPZ. This study demonstrates the novel design of in situ PC-mediated multifunctional liposomes for hypoxia-activated chemotherapy combined with PDT, a promising approach to cancer therapy.
Collapse
Affiliation(s)
- Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Wenyu Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Dermatology, Yanbian University Hospital, Yanji 133000, China
| | - Bowen Zeng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Nuoya Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuangqing Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
33
|
Sun D, Hou H, Feng F, Wu W, Tan J, Xie T, Liu J, Wang J, Qian H, Li J, Xing P. A cohort-based multi-omics identifies nuclear translocation of eIF5B /PD-L1/CD44 complex as the target to overcome Osimertinib resistance of ARID1A-deficient lung adenocarcinoma. Exp Hematol Oncol 2025; 14:3. [PMID: 39773749 PMCID: PMC11705878 DOI: 10.1186/s40164-024-00594-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Osimertinib has emerged as a critical element in the treatment landscape following recent clinical trials. Further investigation into the mechanisms driving resistance to Osimertinib is necessary to address the restricted treatment options and survival advantages that are compromised by resistance in patients with EGFR-mutated lung adenocarcinoma (LUAD). METHODS Spatial transcriptomic and proteomic analyses were utilized to investigate the mechanisms of Osimertinib resistance. Co-IP, MS, RNA-seq, ChIP-seq, RIP-seq, and ATAC-seq were performed in cell lines to further explore the mechanism. To validate the findings, in vitro and in vivo molecular experiments were conducted. RESULTS We found that the ARID1A deficiency results in resistance to Osimertinib by hindering programmed cell death through the EZH2/PTEN/E2F1 axis. This altered axis influences PD-L1 transcription through E2F1-mediated promoter activation and PD-L1 translation via the MDM2/eIF5B/PD-L1 axis. Subsequently, ARID1A deficiency results in increased expression of eIF5B and Importin-β1, promoting PD-L1 nuclear-translocation. The nuclear PD-L1 (nPD-L1) interacts with CD44, leading to nPD-L1 complex formation, activation of the RASGEF1A promoter, initiation of the Ras pathway, and contributing to Osimertinib resistance. Targeting the transcription, translation and nuclear-translocation of PD-L1 using lipid nanoparticles (LNPs) overcomes ARID1A deficiency-induced resistance. CONCLUSION ARID1A deficiency promotes PD-L1 nuclear translocation and induces Osimertinib resistance.
Collapse
Affiliation(s)
- Dantong Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Medical Oncology, Peking University First Hospital, Beijing, 100034, China
| | - Helei Hou
- Department of Oncology, The Affiliated Hospital of Qingdao University, No. 7 Jiaxing Road, Qingdao, 266000, Shandong, China
| | - Feiyue Feng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Weizheng Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jingyu Tan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Tongji Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiayu Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jinsong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Pathology, Peking University People's Hospital, Beijing, 100044, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Junling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
34
|
Fu Q, Zhao X, Hu J, Jiao Y, Yan Y, Pan X, Wang X, Jiao F. mRNA vaccines in the context of cancer treatment: from concept to application. J Transl Med 2025; 23:12. [PMID: 39762875 PMCID: PMC11702060 DOI: 10.1186/s12967-024-06033-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Immuno-oncology has witnessed remarkable advancements in the past decade, revolutionizing the landscape of cancer therapeutics in an encouraging manner. Among the diverse immunotherapy strategies, mRNA vaccines have ushered in a new era for the therapeutic management of malignant diseases, primarily due to their impressive impact on the COVID-19 pandemic. In this comprehensive review, we offer a systematic overview of mRNA vaccines, focusing on the optimization of structural design, the crucial role of delivery materials, and the administration route. Additionally, we summarize preclinical studies and clinical trials to provide valuable insights into the current status of mRNA vaccines in cancer treatment. Furthermore, we delve into a systematic discussion on the significant challenges facing the current development of mRNA tumor vaccines. These challenges encompass both intrinsic and external factors that are closely intertwined with the successful application of this innovative approach. To pave the way for a more promising future in cancer treatments, a deeper understanding of immunological mechanisms, an increasing number of high-quality clinical trials, and a well-established manufacturing platform are crucial. Collaborative efforts between scientists, clinicians, and industry engineers are essential to achieving these goals.
Collapse
Affiliation(s)
- Qiang Fu
- School of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, 264003, P. R. China
| | - Xiaoming Zhao
- Center of Physical Examination, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China
| | - Jinxia Hu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China
| | - Yang Jiao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, P. R. China
| | - Yunfei Yan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China
| | - Xuchen Pan
- Department of Clinical Laboratory & Health Service Training, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China
| | - Xin Wang
- Department of Clinical Laboratory & Health Service Training, Binzhou Medical University Affiliated 970 Hospital of the PLA Joint Logistic Support Force, Yantai, 264002, P. R. China.
| | - Fei Jiao
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, P. R. China.
| |
Collapse
|
35
|
Rampado R, Naidu GS, Karpov O, Goldsmith M, Sharma P, Ezra A, Stotsky L, Breier D, Peer D. Lipid Nanoparticles With Fine-Tuned Composition Show Enhanced Colon Targeting as a Platform for mRNA Therapeutics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408744. [PMID: 39585189 PMCID: PMC11744673 DOI: 10.1002/advs.202408744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/11/2024] [Indexed: 11/26/2024]
Abstract
Lipid Nanoparticles (LNPs) recently emerged as an invaluable RNA delivery platform. With many LNP-based therapeutics in the pre-clinical and clinical pipelines, there is extensive research dedicated to improving LNPs. These efforts focus mainly on the tolerability and transfectability of new ionizable lipids and RNAs, or modulating LNPs biodistribution with active targeting strategies. However, most formulations follow the well-established lipid proportions used in clinically approved products. Nevertheless, investigating the effects of LNPs composition on their biodistribution can expand the toolbox for particle design, leading to improved delivery strategies. Herein, a new LNPs (30-n-LNPs) formulation with increasing amounts of phospholipids is investigated as a possible mRNA delivery system for treating Inflammatory Bowel Diseases. Compared to LNPs with benchmark composition (b-LNPs), n-LNPs containing 30% distearoylphosphatidylcholine (DSPC) are well tolerated following intravenous administration and display natural targeting toward the inflamed colon in dextran sodium sulfate (DSS)-colitis bearing mice, while de-targeting clearing organs such as the liver and spleen. Using interleukin-10-encoding mRNA as therapeutic cargo, n-LNPs demonstrated a reduction of pathological burden in colitis-bearing mice. n-LNPs represent a starting point to further investigate the influence of LNPs composition on systemic biodistribution, ultimately opening new therapeutic modalities in different pathologies.
Collapse
Affiliation(s)
- Riccardo Rampado
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Gonna Somu Naidu
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Olga Karpov
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Meir Goldsmith
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Preeti Sharma
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Assaf Ezra
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Lior Stotsky
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Dor Breier
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Dan Peer
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| |
Collapse
|
36
|
Yao R, Xie C, Xia X. Recent progress in mRNA cancer vaccines. Hum Vaccin Immunother 2024; 20:2307187. [PMID: 38282471 PMCID: PMC10826636 DOI: 10.1080/21645515.2024.2307187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/16/2024] [Indexed: 01/30/2024] Open
Abstract
The research and development of messenger RNA (mRNA) cancer vaccines have gradually overcome numerous challenges through the application of personalized cancer antigens, structural optimization of mRNA, and the development of alternative RNA-based vectors and efficient targeted delivery vectors. Clinical trials are currently underway for various cancer vaccines that encode tumor-associated antigens (TAAs), tumor-specific antigens (TSAs), or immunomodulators. In this paper, we summarize the optimization of mRNA and the emergence of RNA-based expression vectors in cancer vaccines. We begin by reviewing the advancement and utilization of state-of-the-art targeted lipid nanoparticles (LNPs), followed by presenting the primary classifications and clinical applications of mRNA cancer vaccines. Collectively, mRNA vaccines are emerging as a central focus in cancer immunotherapy, offering the potential to address multiple challenges in cancer treatment, either as standalone therapies or in combination with current cancer treatments.
Collapse
Affiliation(s)
- Ruhui Yao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunyuan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
37
|
Gao R, Hu Y, Yuan Q. ADAMTS12 serves as a novel prognostic biomarker and promotes proliferation and invasion in gastric cancer. Discov Oncol 2024; 15:837. [PMID: 39720953 DOI: 10.1007/s12672-024-01724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Gastric cancer (GC) remains a prevalent and aggressive malignancy with a poor prognosis. This study aimed to identify diagnostic and prognostic biomarkers while exploring their potential functions in GC. A total of 598 upregulated and 506 downregulated genes were identified in GC patients. Among these, survival-related differentially expressed genes (DEGs), including ADAMTS12, F5, and VCAN, were highlighted. Pan-cancer analyses revealed their dysregulation across multiple tumor types. A novel prognostic signature, incorporating ADAMTS12 and F5, effectively stratified GC patients into low- and high-risk groups, demonstrating significant differences in overall survival and robust predictive performance. ADAMTS12, strongly associated with advanced clinical stages and poor prognosis, was validated in an independent cohort and exhibited promising diagnostic potential. RT-PCR and western blot analyses confirmed its high expression in GC tissues and cell lines. Functional assays further demonstrated that ADAMTS12 promotes GC cell proliferation and invasion. In summary, this study provides critical insights into the molecular landscape of GC, offering a potential prognostic tool and therapeutic target.
Collapse
Affiliation(s)
- Ruimei Gao
- Department of Gastroenterology, Qingdao Chengyang People's Hospital, Qingdao, China
| | - Yalan Hu
- Department of Anorectal Surgery, Qingdao Eighth People's Hospital, Qingdao, China
| | - Qiuxiang Yuan
- Department of Gastroenterology, Qingdao Chengyang People's Hospital, Qingdao, China.
| |
Collapse
|
38
|
Gehrke L, Gonçalves VDR, Andrae D, Rasko T, Ho P, Einsele H, Hudecek M, Friedel SR. Current Non-Viral-Based Strategies to Manufacture CAR-T Cells. Int J Mol Sci 2024; 25:13685. [PMID: 39769449 PMCID: PMC11728233 DOI: 10.3390/ijms252413685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The successful application of CAR-T cells in the treatment of hematologic malignancies has fundamentally changed cancer therapy. With increasing numbers of registered CAR-T cell clinical trials, efforts are being made to streamline and reduce the costs of CAR-T cell manufacturing while improving their safety. To date, all approved CAR-T cell products have relied on viral-based gene delivery and genomic integration methods. While viral vectors offer high transfection efficiencies, concerns regarding potential malignant transformation coupled with costly and time-consuming vector manufacturing are constant drivers in the search for cheaper, easier-to-use, safer, and more efficient alternatives. In this review, we examine different non-viral gene transfer methods as alternatives for CAR-T cell production, their advantages and disadvantages, and examples of their applications. Transposon-based gene transfer methods lead to stable but non-targeted gene integration, are easy to handle, and achieve high gene transfer rates. Programmable endonucleases allow targeted integration, reducing the potential risk of integration-mediated malignant transformation of CAR-T cells. Non-integrating CAR-encoding vectors avoid this risk completely and achieve only transient CAR expression. With these promising alternative techniques for gene transfer, all avenues are open to fully exploiting the potential of next-generation CAR-T cell therapy and applying it in a wide range of applications.
Collapse
Affiliation(s)
- Leon Gehrke
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Vasco Dos Reis Gonçalves
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Dominik Andrae
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Tamas Rasko
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Patrick Ho
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
- Fraunhofer-Institut für Zelltherapie und Immunologie, Außenstelle Zelluläre Immuntherapie, 97070 Würzburg, Germany
| | - Sabrina R. Friedel
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
39
|
Jiao Y, Yang L, Wang R, Song G, Fu J, Wang J, Gao N, Wang H. Drug Delivery Across the Blood-Brain Barrier: A New Strategy for the Treatment of Neurological Diseases. Pharmaceutics 2024; 16:1611. [PMID: 39771589 PMCID: PMC11677317 DOI: 10.3390/pharmaceutics16121611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The blood-brain barrier (BBB) serves as a highly selective barrier between the blood and the central nervous system (CNS), and its main function is to protect the brain from foreign substances. This physiological property plays a crucial role in maintaining CNS homeostasis, but at the same time greatly limits the delivery of drug molecules to the CNS, thus posing a major challenge for the treatment of neurological diseases. Given that the high incidence and low cure rate of neurological diseases have become a global public health problem, the development of effective BBB penetration technologies is important for enhancing the efficiency of CNS drug delivery, reducing systemic toxicity, and improving the therapeutic outcomes of neurological diseases. This review describes the physiological and pathological properties of the BBB, as well as the current challenges of trans-BBB drug delivery, detailing the structural basis of the BBB and its role in CNS protection. Secondly, this paper reviews the drug delivery strategies for the BBB in recent years, including physical, biological and chemical approaches, as well as nanoparticle-based delivery technologies, and provides a comprehensive assessment of the effectiveness, advantages and limitations of these delivery strategies. It is hoped that the review in this paper will provide valuable references and inspiration for future researchers in therapeutic studies of neurological diseases.
Collapse
Affiliation(s)
- Yimai Jiao
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Luosen Yang
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China;
| | - Rujuan Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Guoqiang Song
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Jingxuan Fu
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China;
| | - Jinping Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Na Gao
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Hui Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| |
Collapse
|
40
|
Shariati A, Khani P, Nasri F, Afkhami H, Khezrpour A, Kamrani S, Shariati F, Alavimanesh S, Modarressi MH. mRNA cancer vaccines from bench to bedside: a new era in cancer immunotherapy. Biomark Res 2024; 12:157. [PMID: 39696625 DOI: 10.1186/s40364-024-00692-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/15/2024] [Indexed: 12/20/2024] Open
Abstract
Harnessing the power of the immune system to target cancer cells is one of the most appealing approaches for cancer therapy. Among these immunotherapies, messenger ribonucleic acid (mRNA) cancer vaccines are worthy of consideration, as they have demonstrated promising results in clinical trials. These vaccines have proven to be safe and well-tolerated. They can be easily mass-produced in a relatively short time and induce a systemic immune response effective against both the primary tumor and metastases. Transcripts encoding immunomodulatory molecules can also be incorporated into the mRNA, enhancing its efficacy. On the other hand, there are some challenges associated with their application, including mRNA instability, insufficient uptake by immune cells, and intrinsic immunogenicity, which can block mRNA translation. Many innovations have been suggested to overcome these obstacles, including structural modification (such as 5' cap modification), optimizing delivery vehicles (especially dendritic cells (DCs) and nanoparticles), and using antigens that can enhance immunogenicity by circumventing tolerance mechanisms. A popular approach is to combine mRNA cancer vaccines with traditional and novel cancer treatments like chemotherapy, radiotherapy, and immune checkpoint blockade (ICB). They are most efficacious when combined with other therapies like ICBs. There is still a long way to go before these vaccines enter the standard of care for cancer patients, but with the incredible pace of development in this field, their clinical application will soon be witnessed. This review highlights the recent advances and challenges of mRNA cancer vaccines. Finally, some of the most prominent clinical applications of these vaccines will be reviewed.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
41
|
Fu T, Zhou B, Li Y, Liu W, Xie Y, Mo Z, Yin F, Wang Y, Fang K, Fang Y, Xiong Z, Yu K, Le A. Innovative Dual mRNA-Lipid Nanoparticle Therapy Targeting CRHBP and CFHR3 for Enhanced Treatment of Hepatocellular Carcinoma. Int J Nanomedicine 2024; 19:13183-13199. [PMID: 39664759 PMCID: PMC11633302 DOI: 10.2147/ijn.s498065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is a deadly disease requiring the identification of new therapeutic targets and strategies. Methods This study identified genes linked to HCC progression via differential analysis. Key genes were identified through univariate and multivariate Cox regression analysis. The biological effects of co-expressed CRHBP and CFHR3 were evaluated in vitro. mRNAs encoding CRHBP and CFHR3 were encapsulated in lipid nanoparticles (LNPs), with the addition of SP94 peptide on the LNPs surface to enhance targeting. The therapeutic efficacy of dual-mRNA LNPs was evaluated in HCC cells and mouse models. Results CRHBP and CFHR3 were closely associated with HCC progression. Low expression of CRHBP (P < 0.01, HR = 1.931 [1.174-3.175]) and CFHR3 (P < 0.05, HR = 1.755 [1.066-2.890]) was identified as a poor prognostic factor for HCC. The risk score model combining CRHBP and CFHR3 demonstrated superior predictive power (P < 0.001, HR = 2.935 [1.768-4.872]). Co-expression of CRHBP and CFHR3 significantly inhibited the malignant biological functions of HCC cells. Treatment with SP94 peptide-modified dual-mRNA LNPs markedly suppressed HCC tumor growth and exhibited excellent biocompatibility and safety. Conclusion Our study proposes a dual-targeted therapeutic strategy for HCC, which may represent a promising treatment approach.
Collapse
Affiliation(s)
- Tianmei Fu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Boxuan Zhou
- Department of Breast Disease Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yingliang Li
- Department of Breast Disease Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Wei Liu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yuankang Xie
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Zhaohong Mo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanchang, People’s Republic of China
| | - Fang Yin
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yu Wang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Kang Fang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yangyang Fang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Ziqing Xiong
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Kuai Yu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Aiping Le
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
42
|
Wu SH, Xiao MC, Liu F, Hong HY, Ding CH, Zhang X, Xie WF. Cell-permeated peptide P-T3H2 inhibits malignancy on hepatocellular carcinoma through stabilizing HNF4α protein. Discov Oncol 2024; 15:752. [PMID: 39638897 PMCID: PMC11621286 DOI: 10.1007/s12672-024-01661-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
OBJECTIVES Hepatocyte nuclear factor 4α (HNF4α) is a key regulator of hepatocyte function and has a strong therapeutic effect on hepatocellular carcinoma (HCC) by inducing the differentiation of hepatoma cell into hepatocytes. Our previous study showed that Tribbles homolog 3 (TRIB3) directly interacts with and promotes the degradation of HNF4α in non-alcoholic fatty liver disease (NAFLD). Disrupting the TRIB3-HNF4α interaction by a cell-permeating peptide, called P-T3H2, stabilized HNF4α protein. This study aimed to assess the anti-tumor impact of P-T3H2 in HCC. METHODS The expression of TRIB3 and HNF4α was evaluated using western blot and immunohistochemistry (IHC). Hepatic functions and cellular senescence of HCC cells were evaluated through periodic acid-Schiff (PAS) staining, acetylated low-density lipoprotein (ac-LDL) uptake and senescence-associated β-galactosidase (SA-β-gal) activity staining, respectively. RNA-Seq analysis was performed to identify differentially expressed genes in Huh7 cells treated with P-T3H2. The impact of P-T3H2 on HCC malignancy was assessed in vitro and in vivo. RESULTS TRIB3 exhibited a negative correlation with HNF4α in both human and mouse HCC tissues. The administration of P-T3H2 significantly inhibited the malignancy of HCC cells. Additionally, P-T3H2 stabilized HNF4α protein and facilitated the restoration of hepatic functions and the cellular senescence in HCC cells. RNA-Seq analysis demonstrated that P-T3H2 enhanced the transcriptional activity of HNF4α in HCC. Furthermore, P-T3H2 effectively suppressed the carcinogenesis and progression of HCC in mice. CONCLUSION P-T3H2 suppressed HCC progression through the stabilization of HNF4α protein and may be a promising therapeutic candidate for clinical application in the treatment of HCC.
Collapse
Affiliation(s)
- Si-Han Wu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Meng-Chao Xiao
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fang Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Huan-Yu Hong
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen-Hong Ding
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
43
|
Mei T, Ye T, Huang D, Xie Y, Xue Y, Zhou D, Wang W, Chen J. Triggering immunogenic death of cancer cells by nanoparticles overcomes immunotherapy resistance. Cell Oncol (Dordr) 2024; 47:2049-2071. [PMID: 39565509 DOI: 10.1007/s13402-024-01009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/21/2024] Open
Abstract
Immunotherapy resistance poses a significant challenge in oncology, necessitating novel strategies to enhance the therapeutic efficacy. Immunogenic cell death (ICD), including necroptosis, pyroptosis and ferroptosis, triggers the release of tumor-associated antigens and numerous bioactive molecules. This release can potentiate a host immune response, thereby overcoming resistance to immunotherapy. Nanoparticles (NPs) with their biocompatible and immunomodulatory properties, are emerging as promising vehicles for the delivery of ICD-inducing agents and immune-stimulatory adjuvants to enhance immune cells tumoral infiltration and augment immunotherapy efficacy. This review explores the mechanisms underlying immunotherapy resistance, and offers an in-depth examination of ICD, including its principles and diverse modalities of cell death that contribute to it. We also provide a thorough overview of how NPs are being utilized to trigger ICD and bolster antitumor immunity. Lastly, we highlight the potential of NPs in combination with immunotherapy to revolutionize cancer treatment.
Collapse
Affiliation(s)
- Ting Mei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ting Ye
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dingkun Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Yuxiu Xie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Ying Xue
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dongfang Zhou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Weimin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Wuhan, 430022, China.
- Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
44
|
Chen BH, Xia QS, Li J, Cai GX, Wang Q. Enhancing the Encapsulation Performances of Liposomes for Amphiphilic Copolymers by Computer Simulations. J Phys Chem B 2024; 128:11481-11491. [PMID: 39504498 DOI: 10.1021/acs.jpcb.4c05650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Liposomes, which encapsulate drugs into an inner aqueous core and demonstrate high drug-loading capacity, have attracted considerable interest in the field of drug delivery. Herein, the encapsulation processes for amphiphilic copolymers within liposomes have been investigated systematically to enhance the encapsulation capacity and optimize the structures using dissipative particle dynamics simulations. The results indicate that the physicochemical properties of lipids, receptors, and amphiphilic copolymers collectively determine the encapsulation behaviors of liposomes. Adjusting the hydrophobic interaction between hydrophobic tails of lipids (receptors) and hydrophobic blocks of copolymers, along with modulating the specific interaction between ligands and the functional head groups of receptors, can lead to various encapsulation capacities. Significantly, a medium hydrophobic interaction strength or a strong specific interaction is conducive to achieving a higher degree of encapsulation for amphiphilic copolymers. Furthermore, varying the key parameters, such as the hydrophobic interaction, the specific interaction, as well as the concentrations of lipids and receptors, can induce seven typical aggregate structures: heterogeneous, fully encapsulated, partially encapsulated, saturated-encapsulated, unsaturated-encapsulated, multilamellar, and column-like structures. The final phase diagrams are also constructed to provide a guideline for designing various structures of liposomes encapsulated with amphiphilic copolymers. These results significantly contribute to the illumination of strategies for the rational construction of the self-assembly system that facilitates the efficient encapsulation of amphiphilic copolymers within the inner aqueous core of liposomes, thereby providing valuable insights into the optimal design of liposome carriers for future biomedical applications.
Collapse
Affiliation(s)
- Bo-Han Chen
- School of Electronic Engineering and Intelligent Manufacturing, Anqing Normal University, Anqing 246133, China
| | - Qiang-Sheng Xia
- School of Electronic Engineering and Intelligent Manufacturing, Anqing Normal University, Anqing 246133, China
| | - Juan Li
- School of Mathematics and Physics, Anqing Normal University, Anqing 246133, China
| | - Gai-Xiang Cai
- School of Mathematics and Physics, Anqing Normal University, Anqing 246133, China
| | - Qiang Wang
- School of Electronic Engineering and Intelligent Manufacturing, Anqing Normal University, Anqing 246133, China
| |
Collapse
|
45
|
Shan X, Cai Y, Zhu B, Zhou L, Sun X, Xu X, Yin Q, Wang D, Li Y. Rational strategies for improving the efficiency of design and discovery of nanomedicines. Nat Commun 2024; 15:9990. [PMID: 39557860 PMCID: PMC11574076 DOI: 10.1038/s41467-024-54265-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The rise of rational strategies in nanomedicine development, such as high-throughput methods and computer-aided techniques, has led to a shift in the design and discovery patterns of nanomedicines from a trial-and-error mode to a rational mode. This transition facilitates the enhancement of efficiency in the preclinical discovery pipeline of nanomaterials, particularly in improving the hit rate of nanomaterials and the optimization efficiency of promising candidates. Herein, we describe a directed evolution mode of nanomedicines driven by data to accelerate the discovery of nanomaterials with high delivery efficiency. Computer-aided design strategies are introduced in detail as one of the cutting-edge directions for the development of nanomedicines. Ultimately, we look forward to expanding the tools for the rational design and discovery of nanomaterials using multidisciplinary approaches. Rational design strategies may potentially boost the delivery efficiency of next-generation nanomedicines.
Collapse
Affiliation(s)
- Xiaoting Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, 264000, China
| | - Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Lingli Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xujie Sun
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Dangge Wang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201260, China.
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, 264000, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China.
| |
Collapse
|
46
|
Shi Y, Shi M, Wang Y, You J. Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct Target Ther 2024; 9:322. [PMID: 39543114 PMCID: PMC11564800 DOI: 10.1038/s41392-024-02002-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
In the last decade, messenger ribonucleic acid (mRNA)-based drugs have gained great interest in both immunotherapy and non-immunogenic applications. This surge in interest can be largely attributed to the demonstration of distinct advantages offered by various mRNA molecules, alongside the rapid advancements in nucleic acid delivery systems. It is noteworthy that the immunogenicity of mRNA drugs presents a double-edged sword. In the context of immunotherapy, extra supplementation of adjuvant is generally required for induction of robust immune responses. Conversely, in non-immunotherapeutic scenarios, immune activation is unwanted considering the host tolerability and high expression demand for mRNA-encoded functional proteins. Herein, mainly focused on the linear non-replicating mRNA, we overview the preclinical and clinical progress and prospects of mRNA medicines encompassing vaccines and other therapeutics. We also highlight the importance of focusing on the host-specific variations, including age, gender, pathological condition, and concurrent medication of individual patient, for maximized efficacy and safety upon mRNA administration. Furthermore, we deliberate on the potential challenges that mRNA drugs may encounter in the realm of disease treatment, the current endeavors of improvement, as well as the application prospects for future advancements. Overall, this review aims to present a comprehensive understanding of mRNA-based therapies while illuminating the prospective development and clinical application of mRNA drugs.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Meixing Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, P. R. China.
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, P. R. China.
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, P. R. China.
| |
Collapse
|
47
|
Chen Z, Shu J, Hu Y, Mei H. Synergistic integration of mRNA-LNP with CAR-engineered immune cells: Pioneering progress in immunotherapy. Mol Ther 2024; 32:3772-3792. [PMID: 39295145 PMCID: PMC11573621 DOI: 10.1016/j.ymthe.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/18/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has emerged as a revolutionary approach in the treatment of malignancies. Despite its remarkable successes, this field continues to grapple with challenges such as scalability, safety concerns, limited therapeutic effect, in vivo persistence, and the need for precise control over CAR expression. In the post-pandemic era of COVID-19 vaccine immunization, the application of messenger RNA (mRNA) encapsulated within lipid nanoparticles (LNPs) has recently garnered significant attention as a potential solution to address these challenges. This review delves into the dynamic landscape of mRNA-LNP technology and its potential implications for CAR-engineered immune cell-based immunotherapy.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Jinhui Shu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
48
|
Li M, Wang Z, Tao J, Jiang H, Yang H, Guo D, Zhao H, He X, Luo S, Jiang X, Yuan L, Xiao L, He H, Yu R, Fang J, Liang T, Mao Z, Xu D, Lu Z. Fructose-1,6-bisphosphatase 1 dephosphorylates and inhibits TERT for tumor suppression. Nat Chem Biol 2024; 20:1505-1513. [PMID: 38538923 DOI: 10.1038/s41589-024-01597-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/01/2024] [Indexed: 04/24/2024]
Abstract
Telomere dysfunction is intricately linked to the aging process and stands out as a prominent cancer hallmark. Here we demonstrate that telomerase activity is differentially regulated in cancer and normal cells depending on the expression status of fructose-1,6-bisphosphatase 1 (FBP1). In FBP1-expressing cells, FBP1 directly interacts with and dephosphorylates telomerase reverse transcriptase (TERT) at Ser227. Dephosphorylated TERT fails to translocate into the nucleus, leading to the inhibition of telomerase activity, reduction in telomere lengths, enhanced senescence and suppressed tumor cell proliferation and growth in mice. Lipid nanoparticle-mediated delivery of FBP1 mRNA inhibits liver tumor growth. Additionally, FBP1 expression levels inversely correlate with TERT pSer227 levels in renal and hepatocellular carcinoma specimens and with poor prognosis of the patients. These findings demonstrate that FBP1 governs cell immortality through its protein phosphatase activity and uncover a unique telomerase regulation in tumor cells attributed to the downregulation or deficiency of FBP1 expression.
Collapse
Affiliation(s)
- Min Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jingjing Tao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Dong Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuxiao He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Shudi Luo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoming Jiang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Li Yuan
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Haiyan He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jing Fang
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
49
|
Fei Y, Yu X, Liu P, Ren H, Wei T, Cheng Q. Simplified Lipid Nanoparticles for Tissue- And Cell-Targeted mRNA Delivery Facilitate Precision Tumor Therapy in a Lung Metastasis Mouse Model. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409812. [PMID: 39390844 DOI: 10.1002/adma.202409812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/26/2024] [Indexed: 10/12/2024]
Abstract
mRNA-based applications have achieved remarkable success in the development of next-generation vaccines and the treatment of diverse liver diseases. Overcoming the challenge of delivering mRNA to extrahepatic tissues, especially specific cells within tissues, is crucial for precision therapy. In this study, a platform is developed for selective mRNA delivery to desired cells within tissues by combining lipid nanoparticle (LNP)-based targeted delivery with mRNA sequence-controlled expression. Through systematic optimization, a three-component LNP platform is developed, enabling targeted mRNA delivery to the lung, liver, and spleen. The incorporation of unique microRNA target sites into the mRNA scaffold further enhances control over protein translation in specific cells within the target tissue. This combined strategy, named SELECT (Simplified LNP with Engineered mRNA for Cell-type Targeting), demonstrates its efficacy in distinguishing mRNA expression between tumor and normal cells based on intracellular microRNA abundance. SELECT encapsulating mRNA encoding a tumor-specific cytotoxic protein, human ELANE, exhibits selective mRNA delivery to tumor lesions and significant inhibition of tumor growth in a mouse model of melanoma lung metastasis. Overall, SELECT has great potential as a new precision tumor treatment approach and also offers promising prospects for other mRNA therapies targeting specific cell types.
Collapse
Affiliation(s)
- Yuan Fei
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Xiaolu Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peiyu Liu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Hongyu Ren
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tuo Wei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, 100871, China
| |
Collapse
|
50
|
Zhou Y, Liao Y, Fan L, Wei X, Huang Q, Yang C, Feng W, Wu Y, Gao X, Shen X, Zhou J, Xia Z, Zhang Z. Lung-Targeted Lipid Nanoparticle-Delivered siUSP33 Attenuates SARS-CoV-2 Replication and Virulence by Promoting Envelope Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406211. [PMID: 39301916 PMCID: PMC11558077 DOI: 10.1002/advs.202406211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/02/2024] [Indexed: 09/22/2024]
Abstract
As a structural protein of SARS-CoV-2, the envelope (E) protein not only plays a key role in the formation of viral particles, but also forms ion channels and has pathogenic functions, including triggering cell death and inflammatory responses. The stability of E proteins is controlled by the host ubiquitin-proteasome system. By screening human deubiquitinases, it is found that ubiquitin-specific protease 33 (USP33) can enhance the stability of E proteins depending on its deubiquitinase activity, thereby promoting viral replication. In the absence of USP33, E proteins are rapidly degraded, leading to a reduced viral load and inflammation. Using lipid nanoparticle (LNP) encapsulation of siUSP33 by adjusting the lipid components (ionizable cationic lipids), siUSP33 is successfully delivered to mouse lung tissues, rapidly reducing USP33 expression in the lungs and maintaining knockdown for at least 14 days, effectively suppressing viral replication and virulence. This method of delivery allows efficient targeting of the lungs and a response to acute infections without long-term USP33 deficiency. This research, based on the deubiquitination mechanism of USP33 on the E protein, demonstrates that LNP-mediated siRNA delivery targeting USP33 plays a role in antiviral and anti-inflammatory responses, offering a novel strategy for the prevention and treatment of SARS-CoV-2.
Collapse
Affiliation(s)
- Yuzheng Zhou
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Yujie Liao
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangsha410083China
| | - Lujie Fan
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
- Guangzhou LaboratoryGuangzhou510700China
| | - Xiafei Wei
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Qiang Huang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Chuwei Yang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Wei Feng
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Yezi Wu
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Xiang Gao
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Xiaotong Shen
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Jian Zhou
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Zanxian Xia
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangsha410083China
- Hunan Key Laboratory of Animal Models for Human DiseasesHunan Key Laboratory of Medical Genetics & Center for Medical GeneticsSchool of Life SciencesCentral South UniversityChangsha410013China
| | - Zheng Zhang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
- Shenzhen Research Center for Communicable Disease Diagnosis and TreatmentChinese Academy of Medical SciencesShenzhen518112China
| |
Collapse
|