1
|
Gong Z, Cheng C, Sun C, Cheng X. Harnessing engineered extracellular vesicles for enhanced therapeutic efficacy: advancements in cancer immunotherapy. J Exp Clin Cancer Res 2025; 44:138. [PMID: 40317075 PMCID: PMC12048990 DOI: 10.1186/s13046-025-03403-w] [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: 02/13/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025] Open
Abstract
Extracellular vesicles (EVs), particularly engineered variants, have emerged as promising tools in cancer immunotherapy due to their inherent ability to modulate immune responses and deliver therapeutic agents with high specificity and minimal toxicity. These nanometer-sized vesicles, which include exosomes (Exos) and other subtypes, naturally participate in intercellular communication and are capable of carrying a diverse range of bioactive molecules, including proteins, lipids, RNAs, and metabolites. Recent advancements in the biogenesis of engineered EVs, such as strategies to modify their surface characteristics and cargo, have significantly expanded their potential as effective vehicles for targeted cancer therapies. Tailoring the contents of EVs, such as incorporating immunomodulatory molecules or gene-editing tools (GETs), has shown promising outcomes in enhancing anti-tumor immunity and overcoming the immunosuppressive tumor microenvironment (TME). Moreover, optimizing delivery mechanisms, through both passive and active targeting strategies, is crucial for improving the clinical efficacy of EV-based therapies. This review provides an overview of recent developments in the engineering of EVs for cancer immunotherapy, focusing on their biogenesis, methods of content customization, and innovations in cargo delivery. Additionally, the review addresses the challenges associated with the clinical translation of EV-based therapies, such as issues related to scalability, safety, and targeted delivery. By offering insights into the current state of the field and identifying key areas for future research, this review aims to advance the application of engineered EVs in cancer treatment.
Collapse
Affiliation(s)
- Zheng Gong
- Department of Radiology, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110004, China
| | - Cheng Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110004, China
| | - Chaonan Sun
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning, 110042, China.
| | - Xiaoli Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|
2
|
Afshar Y, Sharifi N, Kamroo A, Yazdanpanah N, Saleki K, Rezaei N. Implications of glioblastoma-derived exosomes in modifying the immune system: state-of-the-art and challenges. Rev Neurosci 2025; 36:315-325. [PMID: 39528347 DOI: 10.1515/revneuro-2024-0095] [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: 07/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma is a brain cancer with a poor prognosis. Failure of classical chemotherapy and surgical treatments indicates that new therapeutic approaches are needed. Among cell-free options, exosomes are versatile extracellular vesicles (EVs) that carry important cargo across barriers such as the blood-brain barrier (BBB) to their target cells. This makes exosomes an interesting option for the treatment of glioblastoma. Moreover, exosomes can comprise many therapeutic cargos, including lipids, proteins, and nucleic acids, sampled from special intercellular compartments of their origin cell. Cells exposed to various immunomodulatory stimuli can generate exosomes enriched in specific therapeutic molecules. Notably, the secretion of exosomes could modify the immune response in innate and adaptive immune systems. For instance, glioblastoma-associated exosomes (GBex) uptake by macrophages could influence macrophage dynamics (e.g., shifting CD markers expression). Expression of critical immunoregulatory proteins such as cytotoxic T-lymphocyte antigen-1 (CTLA1) and programmed death-1 (PD-1) on GBex indicates the direct crosstalk of these nano-size vesicles with the immune system. The present study reviews the role of exosomes in immune system cells, including B cells, T cells, natural killer (NK) cells, and dendritic cells (DCs), as well as novel technologies in the field.
Collapse
Affiliation(s)
- Yashmin Afshar
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Negin Sharifi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Faculty of Medicine, Tehran Medical Science Branch, Islamic Azad University, Tehran, 1584743311, Iran
| | - Amirhossein Kamroo
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Students' Scientific Research Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
| | - Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Kiarash Saleki
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Student Research Committee, Babol University of Medical Sciences, Babol, 4717647745, Iran
- USERN Office, Babol University of Medical Sciences, Babol, 4717647745, Iran
- Department of E-Learning in Medical Sciences, Faculty of Medical Education and Learning Technologies, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Stockholms, 10316, Sweden
| |
Collapse
|
3
|
Jang J, Shin J, Ahn Y, Kim K, Cho J, Lee WJ, Nam C, Baek MC, Seo D, Yea K. Modular and Nondisturbing Chimeric Adaptor Protein for Surface Chemistry of Small Extracellular Vesicles. ACS NANO 2025; 19:12839-12852. [PMID: 40119814 DOI: 10.1021/acsnano.4c15441] [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: 03/24/2025]
Abstract
Current chemical strategies for modifying the surface of extracellular vesicles (sEVs) often struggle to balance efficient functionalization with preserving structural integrity. Here, we present a modular approach for the surface modification of sEVs using a chimeric adaptor protein (CAP). The CAP was designed with three key features: a SNAP-tag for stable and modular binding, long and rigid linker to enhance spatial accessibility and conjugation efficiency, and the N-terminal sorting domain derived from syntenin to improve CAP expression on the sEV. We established a postsynthetic method to introduce diverse functional molecules onto sEVs, creating a versatile system termed "sEV-X" (where X represents an organic molecule, protein, or nanoparticle). Quantitative analyses at the single-molecule level revealed a linear relationship between CAP expression and the number of conjugated functional molecules, underscoring the importance of steric hindrance mitigation in sEV surface engineering. Moreover, antibody-conjugated sEVs as drug carriers, demonstrated significant tumor-specific delivery and therapeutic efficacy in a tumor-bearing mouse model, underscoring the potential of CAP-expressing sEVs as a customizable therapeutic vesicle. Overall, the CAP technology may serve as a universal platform for advancing the development of sEV-based therapeutics.
Collapse
Affiliation(s)
- Juhee Jang
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jiwon Shin
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Yongdeok Ahn
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Kiwook Kim
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Juhyeong Cho
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Wonhee John Lee
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Chaerin Nam
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| |
Collapse
|
4
|
Chen Q, Jin J, Li P, Wang X, Wang Q. Navigating Glioma Complexity: The Role of Abnormal Signaling Pathways in Shaping Future Therapies. Biomedicines 2025; 13:759. [PMID: 40149733 PMCID: PMC11940491 DOI: 10.3390/biomedicines13030759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Gliomas are a type of highly heterogeneous and invasive central nervous system tumor. Traditional treatment methods have limited efficacy, and the prognosis for patients remains poor. Recent studies have revealed the crucial roles of several abnormal signaling pathways in the pathogenesis of gliomas, including the Receptor Tyrosine Kinase/Rat Sarcoma Virus Oncogene/Phosphatidylinositol-3-Kinase (RTK/RAS/PI3K) pathway, the Wingless-Related Integration Site/β-Catenin (Wnt/β-Catenin) pathway, the Hippo/YAP (Hippo/Yes-associated protein) pathway, and the Slit/Robo (Slit Guidance Ligands/Roundabout) signaling pathway. These pathways play extremely vital roles in tumor proliferation, invasion, and treatment resistance. This article comprehensively and systematically reviews the molecular mechanisms of these signaling pathways, deeply summarizing the research progress of various treatment strategies, including targeted inhibitors, gene therapy, and nanomedicine against them. Moreover, the combination of targeted therapy and personalized treatment regimens is expected to overcome the current treatment bottleneck and provide a more favorable survival prognosis for glioblastoma patients.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Pharmacy, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Jin Jin
- Department of Rehabilitation, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Pian Li
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Xiuping Wang
- Department of Pharmacy, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Qianyan Wang
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| |
Collapse
|
5
|
Ren X, Wang S, Teng Y, Zheng S, Li F, Wang C, Wu L, Zhang J. Engineered extracellular vesicles loaded in boronated cyclodextrin framework for pulmonary delivery. Carbohydr Polym 2025; 352:123160. [PMID: 39843065 DOI: 10.1016/j.carbpol.2024.123160] [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: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 01/24/2025]
Abstract
Extracellular vesicles (EVs) are promising therapeutic carriers for their ideal nano-size and intrinsic biocompatibility, while rapid clearance and limited targeting ability are the major setbacks of EVs. With minimal absorption into the systemic circulation, inhalation for pulmonary disease therapy minimizes off-target toxicity to other organs and offers a safe and effective treatment for respiratory disorders. Herein, a nano-grid carrier made of boronated cyclodextrin framework (BCF) was prepared for pH/H2O2 responsive release of EVs. A novel design of cyclo (Arg-Gly-Asp-D-Tyr-Lys) peptide (RGD)-modified milk-derived EVs (mEVs) loaded in the BCF particles (RGD-mEVs@BCF) was developed for pulmonary delivery. The results indicated that RGD-mEVs showed superior anti-inflammatory activity in contrast with mEVs in vitro. BCF was able to capture and protect RGD-mEVs, which showed extended-release profiles and responsiveness. Pulmonary administration of RGD-mEVs@BCF showed favorable biocompatibility in rats. Taken together, RGD-mEVs@BCF features biocompatibility and pH-responsive mEVs release as a therapeutic platform for pulmonary delivery of drugs to treat lung diseases, especially for inflammatory diseases.
Collapse
Affiliation(s)
- Xiaohong Ren
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Siwen Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yupu Teng
- Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shiyu Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Feng Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Caifen Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Li Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Jiwen Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Shenyang Pharmaceutical University, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Liu M, Wang Y, Zhang Y, Hu D, Tang L, Zhou B, Yang L. Landscape of small nucleic acid therapeutics: moving from the bench to the clinic as next-generation medicines. Signal Transduct Target Ther 2025; 10:73. [PMID: 40059188 PMCID: PMC11891339 DOI: 10.1038/s41392-024-02112-8] [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: 07/17/2024] [Revised: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 03/17/2025] Open
Abstract
The ability of small nucleic acids to modulate gene expression via a range of processes has been widely explored. Compared with conventional treatments, small nucleic acid therapeutics have the potential to achieve long-lasting or even curative effects via gene editing. As a result of recent technological advances, efficient small nucleic acid delivery for therapeutic and biomedical applications has been achieved, accelerating their clinical translation. Here, we review the increasing number of small nucleic acid therapeutic classes and the most common chemical modifications and delivery platforms. We also discuss the key advances in the design, development and therapeutic application of each delivery platform. Furthermore, this review presents comprehensive profiles of currently approved small nucleic acid drugs, including 11 antisense oligonucleotides (ASOs), 2 aptamers and 6 siRNA drugs, summarizing their modifications, disease-specific mechanisms of action and delivery strategies. Other candidates whose clinical trial status has been recorded and updated are also discussed. We also consider strategic issues such as important safety considerations, novel vectors and hurdles for translating academic breakthroughs to the clinic. Small nucleic acid therapeutics have produced favorable results in clinical trials and have the potential to address previously "undruggable" targets, suggesting that they could be useful for guiding the development of additional clinical candidates.
Collapse
Affiliation(s)
- Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yibing Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Xiong Y, Sun M, Yang Q, Zhang W, Song A, Tan Y, Mao J, Liu G, Xue P. Nanoparticle-based drug delivery systems to modulate tumor immune response for glioblastoma treatment. Acta Biomater 2025; 194:38-57. [PMID: 39884522 DOI: 10.1016/j.actbio.2025.01.050] [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: 10/16/2024] [Revised: 12/28/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
Glioblastoma (GBM) is a primary central nervous system neoplasm, characterized by a grim prognosis and low survival rates. This unfavorable therapeutic outcome is partially attributed to the inadequate immune infiltration and an immunosuppressive microenvironment, which compromises the effectiveness of conventional radiotherapy and chemotherapy. To this end, precise modulation of cellular dynamics in the immune system has emerged as a promising approach for therapeutic intervention. The advent of nanoparticle-based therapies has revolutionized cancer treatment and provided highly effective options. Consequently, various strategically designed nano-delivery platforms have been established to promote the efficacy of immune therapy against GBM. This review delves into the recent advancements in nano-based delivery systems that are designed to modulate immune cells in GBM microenvironment, and explores their multifaceted mechanisms, including the blockade of immune checkpoints, the restraint of immunosuppressive cells, the coordination of tumor-associated macrophages, the activation of innate immune cells, and the stimulation of adaptive immunity. Collectively, this summary not only advances the comprehension involved in modulating antitumor immune responses in GBM, but also paves the way for the development of innovative therapeutic strategies to conquer GBM. STATEMENT OF SIGNIFICANCE: Glioblastoma (GBM) is the most lethal brain tumor, with a median survival rate of merely 12-16 months after diagnosis. Despite surgical, radiation and chemotherapy treatments, the two-year survival rate for GBM patients is less than 10 %. The treatment of GBM is challenging mainly because several issues associated with the GBM microenvironment have not yet been resolved. Most recently, novel drug delivery approaches, based on the clear understanding of the intrinsic properties of GBM, have shown promise in overcoming some of the obstacles. In particular, taking account of the highly immunosuppressive tumor microenvironment in GBM, recent advancements in nano-based delivery systems are put forward to stimulate immune cells in GBM and unravel their multifaceted mechanisms. This review summarizes the latest nanoparticle-based drug delivery systems to modulate tumor immune response for glioblastoma treatment. Moreover, the development trends and challenges of nanoparticle-based drug delivery systems in modulating the immunity of GBM are predicted, which may facilitate widespread regimens springing up for successfully treating GBM.
Collapse
Affiliation(s)
- Yongqi Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Maoyuan Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qinhao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wenli Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Anchao Song
- College of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang 550000, China
| | - Jinning Mao
- Health Medical Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Peng Xue
- School of Materials and Energy, Southwest University, Chongqing 400715, China; Yibin Academy of Southwest University, Yibin 644005, China.
| |
Collapse
|
8
|
Grzegorzewski J, Michalak M, Wołoszczuk M, Bulicz M, Majchrzak-Celińska A. Nanotherapy of Glioblastoma-Where Hope Grows. Int J Mol Sci 2025; 26:1814. [PMID: 40076445 PMCID: PMC11898975 DOI: 10.3390/ijms26051814] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Localization in the central nervous system, diffuse growth, the presence of stem cells, and numerous resistance mechanisms, all make glioblastoma (GBM) an incurable tumor. The standard treatment of GBM consisting of surgery; radio- and chemotherapy with temozolomide provides insufficient therapeutic benefit and needs to be updated with effective modern solutions. One of the most promising and intensively explored therapeutic approaches against GBM is the use of nanotherapy. The first, and so far only, nanoparticle-based therapy approved for GBM treatment is NanoThermTM. It is based on iron oxide nanoparticles and the thermal ablation of the tumor with a magnetic field. Numerous other types of nanotherapies are being evaluated, including polymer and lipid-based nanoformulations, nanodiscs, dendrimers, and metallic, silica, or bioderived nanoparticles, among others. The advantages of these nanoscale drug carriers include improved penetration across the blood-brain barrier, targeted drug delivery, biocompatibility, and lower systemic toxicity, while major problems with their implementation involve scaling up their production and high costs. Nevertheless, taking all the impressive benefits of nanotherapies into consideration, it seems obvious that the combined effort of the scientific world will need to be taken to tackle these challenges and implement these novel therapies into clinics, giving hope that the battle against GBM can finally be won.
Collapse
Affiliation(s)
- Jan Grzegorzewski
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Maciej Michalak
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Maria Wołoszczuk
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Magdalena Bulicz
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Aleksandra Majchrzak-Celińska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| |
Collapse
|
9
|
Zemanek T, Danisovic L, Nicodemou A. Exosomes and solid cancer therapy: where are we now? Med Oncol 2025; 42:77. [PMID: 39961904 PMCID: PMC11832697 DOI: 10.1007/s12032-025-02626-3] [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: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Cancer immunotherapy has revolutionized oncology, offering new hope for patients with previously incurable cancers. However, solid tumors remain a significant challenge due to immune evasion, therapeutic resistance, and the immunosuppressive tumor microenvironment. Exosomes, a specialized subset of extracellular vesicles, have emerged as promising tools in cancer therapy owing to their unique role in intercellular communication and immune modulation. These vesicles transport antigens, major histocompatibility complex (MHC) molecules, and immune-modulatory cargo, positioning them as potential platforms for cancer vaccines, drug delivery systems, and combinatorial therapies. Advances in engineered exosomes have improved drug bioavailability, tumor targeting, and immune stimulation, showcasing their potential in personalized medicine. This review highlights their multifaceted role in the tumor microenvironment, and their mechanisms of action in solid cancer therapy. Additionally, we discuss emerging strategies to overcome clinical and technical hurdles, paving the way for novel and effective cancer treatments.
Collapse
Affiliation(s)
- Tomas Zemanek
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- GAMMA - ZA s.r.o., Trencin, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Andreas Nicodemou
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
- GAMMA - ZA s.r.o., Trencin, Slovakia.
| |
Collapse
|
10
|
Sun B, Li R, Ji N, Liu H, Wang H, Chen C, Bai L, Su J, Chen J. Brain-targeting drug delivery systems: The state of the art in treatment of glioblastoma. Mater Today Bio 2025; 30:101443. [PMID: 39866779 PMCID: PMC11759563 DOI: 10.1016/j.mtbio.2025.101443] [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: 08/25/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant brain tumor, characterized by a high mortality rate and a poor prognosis. The blood-brain barrier (BBB) and the blood-tumor barrier (BTB) present significant obstacles to the efficacy of tumor-targeted pharmacotherapy, thereby impeding the therapeutic potential of numerous candidate drugs. Targeting delivery of adequate doses of drug across the BBB to treat GBM has become a prominent research area in recent years. This emphasis has driven the exploration and evaluation of diverse technologies for GBM pharmacotherapy, with some already undergoing clinical trials. This review provides a thorough overview of recent advancements and challenges in targeted drug delivery for GBM treatment. It specifically emphasizes systemic drug administration strategies to assess their potential and limitations in GBM treatment. Furthermore, this review highlights promising future research directions in the development of intelligent drug delivery systems aimed at overcoming current challenges and enhancing therapeutic efficacy against GBM. These advancements not only support foundational research on targeted drug delivery systems for GBM but also offer methodological approaches for future clinical applications.
Collapse
Affiliation(s)
- Bo Sun
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Rong Li
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongxiang Wang
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chao Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
11
|
Maksoud S, Schweiger MW, Tabet EI, Xiao T, Hokayem JE, Zinter M, Carvalho LA, Breyne K, Noske DP, Chiocca EA, Tannous BA. Arming AAV9 with a Single-Chain Fragment Variable Antibody Against PD-1 for Systemic Glioblastoma Therapy. Mol Neurobiol 2025; 62:2617-2625. [PMID: 39138760 PMCID: PMC11772126 DOI: 10.1007/s12035-024-04406-y] [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: 01/04/2024] [Accepted: 07/28/2024] [Indexed: 08/15/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive brain cancer with a low survival rate, prompting the exploration of novel therapeutic strategies. Immune checkpoint inhibitors have shown promise in cancer treatment but are associated with immune-related toxicities and brain penetration. Here, we present a targeted approach using an adeno-associated virus serotype 9 (AAV9) to systemically deliver a single-chain fragment variable antibody against PD-1 (scFv-PD-1) into the tumor microenvironment (TME). Single-cell RNA sequencing analysis revealed robust PD-1 expression in GBM TME, predominantly on T cells. AAV9-scFv-PD-1 expressed and secreted scFv-PD-1, which effectively binds to PD-1. Systemic administration of AAV9-scFv-PD-1 in an immunocompetent GBM mouse model resulted in a robust cytolytic T-cell activation at the tumor site, marked by accumulation of IFN-γ and Granzyme B, leading to a significant reduction in tumor growth. Importantly, AAV9-scFv-PD-1 treatment conferred a survival benefit, highlighting its therapeutic potential. This study demonstrates the feasibility of systemically delivered AAV9-mediated local expression of scFv-PD-1 for targeted immunotherapy in GBM and warrants further investigation for clinical translation.
Collapse
Affiliation(s)
- Semer Maksoud
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA.
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA.
- Present Address: Early Oncology R&D, ICC, AstraZeneca, Waltham, MA, 02451, USA.
| | - Markus W Schweiger
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA
- Department of Neurosurgery, Amsterdam , UMC Location Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Brain Tumor Center and Liquid Biopsy Center, 1081 HV, Amsterdam, the Netherlands
| | - Elie I Tabet
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA
| | - Tianhe Xiao
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA
| | - Joelle El Hokayem
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA
| | - Max Zinter
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA
| | - Litia A Carvalho
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA
| | - Koen Breyne
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA
| | - David P Noske
- Department of Neurosurgery, Amsterdam , UMC Location Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Brain Tumor Center and Liquid Biopsy Center, 1081 HV, Amsterdam, the Netherlands
| | | | - Bakhos A Tannous
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA.
- Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA.
- Present Address: Early Oncology R&D, ICC, AstraZeneca, Waltham, MA, 02451, USA.
| |
Collapse
|
12
|
Wen X, Hao Z, Yin H, Min J, Wang X, Sun S, Ruan G. Engineered Extracellular Vesicles as a New Class of Nanomedicine. CHEM & BIO ENGINEERING 2025; 2:3-22. [PMID: 39975802 PMCID: PMC11835263 DOI: 10.1021/cbe.4c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 02/21/2025]
Abstract
Extracellular vesicles (EVs) are secreted from biological cells and contain many molecules with diagnostic values or therapeutic functions. There has been great interest in academic and industrial communities to utilize EVs as tools for diagnosis or therapeutics. In addition, EVs can also serve as delivery vehicles for therapeutic molecules. An indicator of the enormous interest in EVs is the large number of review articles published on EVs, with the focus ranging from their biology to their applications. An emerging trend in EV research is to produce and utilize "engineered EVs", which are essentially the enhanced version of EVs. EV engineering can be conducted by cell culture condition control, genetic engineering, or chemical engineering. Given their nanometer-scale sizes and therapeutic potentials, engineered EVs are an emerging class of nanomedicines. So far, an overwhelming majority of the research on engineered EVs is preclinical studies; there are only a very small number of reported clinical trials. This Review focuses on engineered EVs, with a more specific focus being their applications in therapeutics. The various approaches to producing engineered EVs and their applications in various diseases are reviewed. Furthermore, in vivo imaging of EVs, the mechanistic understandings, and the clinical translation aspects are discussed. The discussion is primarily on preclinical studies while briefly mentioning the clinical trials. With continued interdisciplinary research efforts from biologists, pharmacists, physicians, bioengineers, and chemical engineers, engineered EVs could become a powerful solution for many major diseases such as neurological, immunological, and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaowei Wen
- Institute
of Analytical Chemistry and Instrument for Life Science, The Key Laboratory
of Biomedical Information Engineering of Ministry of Education, School
of Life Science and Technology, Xi’an
Jiaotong University, Xi’an, China 710049
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Zerun Hao
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Haofan Yin
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Jie Min
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Xueying Wang
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Sihan Sun
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Gang Ruan
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| |
Collapse
|
13
|
He M, Chen S, Yu H, Fan X, Wu H, Wang Y, Wang H, Yin X. Advances in nanoparticle-based radiotherapy for cancer treatment. iScience 2025; 28:111602. [PMID: 39834854 PMCID: PMC11743923 DOI: 10.1016/j.isci.2024.111602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Radiotherapy has long been recognized as an effective conventional approach in both clinical and scientific research, primarily through mechanisms involving DNA destruction or the generation of reactive oxygen species to target tumors. However, significant challenges persist, including the unavoidable damage to normal tissues and the development of radiation resistance. As a result, nanotechnology-based radiotherapy has garnered considerable attention for its potential to enhance precision in irradiation, improve radiosensitization, and achieve therapeutic advancements. Importantly, radiotherapy alone frequently falls short of fully eradicating tumors. Consequently, to augment the efficacy of radiotherapy, it is often integrated with other therapeutic strategies. This review elucidates the mechanisms of radiotherapy sensitization based on diverse nanoparticles. Typically, radiotherapy is sensitized through augmenting reactive oxygen species production, targeted radiotherapy, hypoxia relief, enhancement of antitumor immune microenvironment, and G2/M cell cycle arrest. Moreover, the incorporation of nanoparticle-based anti-tumor strategies with radiotherapy markedly enhances the current state of radiotherapy. Additionally, a compilation of clinical trials utilizing nano-radioenhancers is presented. Finally, future prospects for clinical translation in this field are thoroughly examined.
Collapse
Affiliation(s)
- Meijuan He
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shixiong Chen
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Hongwei Yu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xuhui Fan
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hong Wu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yihui Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Han Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
- Jiading Branch of Shanghai General Hospital, Shanghai 201803, China
| | - Xiaorui Yin
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
14
|
Thakur A, Rai D. Global requirements for manufacturing and validation of clinical grade extracellular vesicles. THE JOURNAL OF LIQUID BIOPSY 2024; 6:100278. [PMID: 40027307 PMCID: PMC11863704 DOI: 10.1016/j.jlb.2024.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 03/05/2025]
Abstract
Extracellular vesicles (EVs) are nanovesicles released from different cell types from biofluids such as blood, urine, and cerebrospinal fluid. They vary in size and biomarkers, and their biogenesis pathways allow them to be divided into three major types: exosomes, micro-vesicles, and apoptotic bodies. EVs have been studied in the context of diagnosis and therapeutic intervention of various pathological conditions such as cancer, neurodegenerative diseases, and pulmonary diseases. However, the production of EV-based therapeutics can be affected by the source, heterogeneity, or disease, raising questions about the manufacturing and validation of EVs of clinical grade and their scope regarding good manufacturing practice (GMP) in the industry. To address this, we have discussed the state-of-the-art requirements for EV production that must occur in a GMP-compliant environment with a reliable and traceable source. Additionally, EVs' homogeneity and the therapeutics' purity and stability must be analyzed and validated. Quality control measures must also be established to ensure the safety and efficacy of EVs. In conclusion, these considerations must be weighed carefully when manufacturing and validating EVs of clinical grade to ensure their safety and efficacy for therapeutic use.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Deepika Rai
- Smidt Heart Institute, Cedars-Sinai Medical Centre, Los Angeles, CA, United States
| |
Collapse
|
15
|
Chen J, Cui J, Jiao B, Zheng Z, Yu H, Wang H, Zhang G, Lai S, Gan Z, Yu Q. Biomimetic Nanosensitizer Potentiates Efficient Glioblastoma Gene-Radiotherapy through Synergistic Hypoxia Mitigation and PLK1 Silencing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58241-58261. [PMID: 39287499 DOI: 10.1021/acsami.4c11566] [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: 09/19/2024]
Abstract
Postoperative radiotherapy currently stands as the cornerstone of glioblastoma (GBM) treatment. Nevertheless, low-dose radiotherapy has been proven ineffective for GBM, due to hypoxia in the GBM microenvironment, which renders the resistance to radiation-induced cell death. Moreover, the overexpression of the PLK1 gene in glioma cells enhances GBM proliferation, invasion, metastasis, and resistance to radiation. This study introduced a hybrid membrane-camouflaged biomimetic lipid nanosensitizer (CNL@miPA), which efficiently encapsulated gold nanoclusters (PA) and miR-593-5p by a chimeric membrane derived from lipids, cancer cells, and natural killer cells. CNL@miPA exhibited exceptional blood-brain barrier and tumor tissue penetration, effectively ameliorating hypoxia and synergizing with radiotherapy. By enabling prolonged miRNA circulation in the bloodstream and achieving high enrichment at the tumor site, CNL@miPA significantly suppressed tumor growth in combination treatment, thereby significantly extending the survival period of treated mice. Overall, the developed biomimetic nanosensitizer represented an efficient and multifunctional targeted delivery system, offering a novel strategy for gene-radiotherapy of GBM.
Collapse
Affiliation(s)
- Jiawei Chen
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiajunzi Cui
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Binbin Jiao
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Ziyan Zheng
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Haiwang Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hanbing Wang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Guan Zhang
- Department of Urology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shicong Lai
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
- The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China
| | - Zhihua Gan
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qingsong Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
16
|
Han T, Hao Q, Chao T, Sun Q, Chen Y, Gao B, Guan L, Ren W, Zhou X. Extracellular vesicles in cancer: golden goose or Trojan horse. J Mol Cell Biol 2024; 16:mjae025. [PMID: 38796692 PMCID: PMC11540518 DOI: 10.1093/jmcb/mjae025] [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: 01/29/2024] [Revised: 04/16/2024] [Accepted: 05/24/2024] [Indexed: 05/28/2024] Open
Abstract
Intercellular communication can be mediated by direct cell-to-cell contact and indirect interactions through secretion of soluble chemokines, cytokines, and growth factors. Extracellular vesicles (EVs) have emerged as important mediators of cell-to-cell and cell-to-environment communications. EVs from tumor cells, immune cells, and stromal cells can remodel the tumor microenvironment and promote cancer cell survival, proliferation, metastasis, immune evasion, and therapeutic resistance. Most importantly, EVs as natural nanoparticles can be manipulated to serve as a potent delivery system for targeted cancer therapy. EVs can be engineered or modified to improve their ability to target tumors and deliver therapeutic substances, such as chemotherapeutic drugs, nucleic acids, and proteins, for the treatment of cancer. This review provides an overview of the biogenesis and recycling of EVs, discusses their roles in cancer development, and highlights their potential as a delivery system for targeted cancer therapy.
Collapse
Affiliation(s)
- Tao Han
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China
| | - Qian Hao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qinggang Sun
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Yitian Chen
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Bo Gao
- Umibio Co. Ltd, Shanghai 201210, China
| | - Liping Guan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Wenjie Ren
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang 453003, China
| | - Xiang Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Zhang H, Zhu W, Pan W, Wan X, Li N, Tang B. Recent advances in spatio-temporally controllable systems for management of glioma. Asian J Pharm Sci 2024; 19:100954. [PMID: 39483717 PMCID: PMC11525460 DOI: 10.1016/j.ajps.2024.100954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 11/03/2024] Open
Abstract
Malignant glioma remains one of the most aggressive intracranial tumors with devastating clinical outcomes despite the great advances in conventional treatment approaches, including surgery and chemotherapy. Spatio-temporally controllable approaches to glioma are now being actively investigated due to the preponderance, including spatio-temporal adjustability, minimally invasive, repetitive properties, etc. External stimuli can be readily controlled by adjusting the site and density of stimuli to exert the cytotoxic on glioma tissue and avoid undesired injury to normal tissues. It is worth noting that the removability of external stimuli allows for on-demand treatment, which effectively reduces the occurrence of side effects. In this review, we highlight recent advancements in drug delivery systems for spatio-temporally controllable treatments of glioma, focusing on the mechanisms and design principles of sensitizers utilized in these controllable therapies. Moreover, the potential challenges regarding spatio-temporally controllable therapy for glioma are also described, aiming to provide insights into future advancements in this field and their potential clinical applications.
Collapse
Affiliation(s)
- Huiwen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Wanqi Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Xiuyan Wan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
- Laoshan Laboratory, Qingdao 266237, China
| |
Collapse
|
18
|
Liu M, Li T, Zhao M, Qian C, Wang R, Liu L, Xiao Y, Xiao H, Tang X, Liu H. Nanoradiosensitizers in glioblastoma treatment: recent advances and future perspectives. Nanomedicine (Lond) 2024; 19:2229-2249. [PMID: 39311492 PMCID: PMC11487349 DOI: 10.1080/17435889.2024.2395238] [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: 06/12/2024] [Accepted: 08/19/2024] [Indexed: 10/16/2024] Open
Abstract
Glioblastoma (GBM), a highly invasive type of brain tumor located within the central nervous system, manifests a median survival time of merely 14.6 months. Radiotherapy kills tumor cells through focused high-energy radiation and has become a crucial treatment strategy for GBM, especially in cases where surgical resection is not viable. However, the presence of radioresistant tumor cells limits its clinical effectiveness. Radioresistance is a key factor of treatment failure, prompting the development of various therapeutic strategies to overcome this challenge. With the rapid development of nanomedicine, nanoradiosensitizers provide a novel approach to enhancing the effectiveness of radiotherapy. In this review, we discuss the reasons behind GBM radio-resistance and the mechanisms of radiotherapy sensitization. Then we summarize the primary types of nanoradiosensitizers and recent progress in their application for the radiosensitization of GBM. Finally, we elucidate the factors influencing their practical implementation, along with the challenges and promising prospects associated with multifunctional nanoradiosensitizers.
Collapse
Affiliation(s)
- Mingxi Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Taiping Li
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Mengjie Zhao
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chunfa Qian
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Ran Wang
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Xiao
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Xianglong Tang
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Liu Y, Li M, Gu J, Huang H, Xie H, Yu C, Roy S, Chen X, Kuang T, Zhang Y, Jiang S, Guo B. Engineering of exosome-liposome hybrid-based theranostic nanomedicines for NIR-II fluorescence imaging-guided and targeted NIR-II photothermal therapy of subcutaneous glioblastoma. Colloids Surf B Biointerfaces 2024; 245:114258. [PMID: 39303384 DOI: 10.1016/j.colsurfb.2024.114258] [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: 07/24/2024] [Revised: 08/27/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Exosome-liposome hybrid-based vehicles (ELV) are promising carriers for cancer treatment, but there are rare efficient theranostic probes to label their lipid bilayer membrane for precisely tracing biodistribution and execute potent therapy. As both fluorescence imaging and photothermal therapy in the second near-infrared window (NIR-II) has intrinsically deep penetration and high efficacy to ablate tumors, herein the design and synthesis of lipophilic NIR-II cyanine dyes with strong donor strength is reported to label lipid bilayer membrane of ELV for NIR-II fluorescence image-guided and targeted NIR-II photothermal treatment of subcutaneous glioblastoma. Via lipid film hydration and subsequent extrusion method, the synthesized ELV (NIR-C12-EL) is formulated with NIR-C12 labeling, cyclic arginylglycylaspartic acid decoration, liposomal PEGylation, and biological exosome function. NIR-C12-EL exhibits excellent colloidal stability, good biocompatibility, strong light harvesting capability, high NIR-II photoconversion efficiency (62.28 %), and targeting capability to diagnose and ablate tumors, which together contribute to the extended life-span of the mice treatment with NIR-C12-EL and continuous 1064 nm laser irradiation. This study provides insight into not only designing of lipophilic NIR-II fluorescence probes for labeling of exosome-liposome hybrid-based vehicles but also the engineering of theranostic nanoplatforms for precise treatment of glioblastoma.
Collapse
Affiliation(s)
- Yue Liu
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Menlong Li
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China
| | - Haiyan Huang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Hui Xie
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu 610041, China
| | - Chen Yu
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Xin Chen
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Ting Kuang
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yinghe Zhang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shengwei Jiang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China.
| |
Collapse
|
20
|
Kang K, Lin X, Chen P, Liu H, Liu F, Xiong W, Li G, Yi M, Li X, Wang H, Xiang B. T cell exhaustion in human cancers. Biochim Biophys Acta Rev Cancer 2024; 1879:189162. [PMID: 39089484 DOI: 10.1016/j.bbcan.2024.189162] [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: 01/30/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
T cell exhaustion refers to a progressive state in which T cells become functionally impaired due to sustained antigenic stimulation, which is characterized by increased expression of immune inhibitory receptors, but weakened effector functions, reduced self-renewal capacity, altered epigenetics, transcriptional programme and metabolism. T cell exhaustion is one of the major causes leading to immune escape of cancer, creating an environment that supports tumor development and metastatic spread. In addition, T cell exhaustion plays a pivotal role to the efficacy of current immunotherapies for cancer. This review aims to provide a comprehensive view of roles of T cell exhaustion in cancer development and progression. We summerized the regulatory mechanisms that involved in T cell exhaustion, including transcription factors, epigenetic and metabolic reprogramming events, and various microenvironmental factors such as cytokines, microorganisms, and tumor autocrine substances. The paper also discussed the challenges posed by T cell exhaustion to cancer immunotherapies, including immune checkpoint blockade (ICB) therapies and chimeric antigen receptor T cell (CAR-T) therapy, highlightsing the obstacles encountered in ICB therapies and CAR-T therapies due to T cell exhaustion. Finally, the article provides an overview of current therapeutic options aimed to reversing or alleviating T cell exhaustion in ICB and CAR-T therapies. These therapeutic approaches seek to overcome T cell exhaustion and enhance the effectiveness of immunotherapies in treating tumors.
Collapse
Affiliation(s)
- Kuan Kang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Xin Lin
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Pan Chen
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Huai Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Feng Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Wei Xiong
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Guiyuan Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Mei Yi
- Department of Dermatology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Infammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| | - Hui Wang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.
| | - Bo Xiang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China; FuRong Laboratory, Changsha 410078, Hunan, China.
| |
Collapse
|
21
|
Mu Y, Zhang Z, Zhou H, Ma L, Wang DA. Applications of nanotechnology in remodeling the tumour microenvironment for glioblastoma treatment. Biomater Sci 2024; 12:4045-4064. [PMID: 38993162 DOI: 10.1039/d4bm00665h] [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: 07/13/2024]
Abstract
With the increasing research and deepening understanding of the glioblastoma (GBM) tumour microenvironment (TME), novel and more effective therapeutic strategies have been proposed. The GBM TME involves intricate interactions between tumour and non-tumour cells, promoting tumour progression. Key therapeutic goals for GBM treatment include improving the immunosuppressive microenvironment, enhancing the cytotoxicity of immune cells against tumours, and inhibiting tumour growth and proliferation. Consequently, remodeling the GBM TME using nanotechnology has emerged as a promising approach. Nanoparticle-based drug delivery enables targeted delivery, thereby improving treatment specificity, facilitating combination therapies, and optimizing drug metabolism. This review provides an overview of the GBM TME and discusses the methods of remodeling the GBM TME using nanotechnology. Specifically, it explores the application of nanotechnology in ameliorating immune cell immunosuppression, inducing immunogenic cell death, stimulating, and recruiting immune cells, regulating tumour metabolism, and modulating the crosstalk between tumours and other cells.
Collapse
Affiliation(s)
- Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
22
|
Yan W, Cao Y, Yin Q, Li Y. Biomimetic Nucleic Acid Drug Delivery Systems for Relieving Tumor Immunosuppressive Microenvironment. Pharmaceutics 2024; 16:1028. [PMID: 39204373 PMCID: PMC11360391 DOI: 10.3390/pharmaceutics16081028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Immunotherapy combats tumors by enhancing the body's immune surveillance and clearance of tumor cells. Various nucleic acid drugs can be used in immunotherapy, such as DNA expressing cytokines, mRNA tumor vaccines, small interfering RNAs (siRNA) knocking down immunosuppressive molecules, and oligonucleotides that can be used as immune adjuvants. Nucleic acid drugs, which are prone to nuclease degradation in the circulation and find it difficult to enter the target cells, typically necessitate developing appropriate vectors for effective in vivo delivery. Biomimetic drug delivery systems, derived from viruses, bacteria, and cells, can protect the cargos from degradation and clearance, and deliver them to the target cells to ensure safety. Moreover, they can activate the immune system through their endogenous activities and active components, thereby improving the efficacy of antitumor immunotherapeutic nucleic acid drugs. In this review, biomimetic nucleic acid delivery systems for relieving a tumor immunosuppressive microenvironment are introduced. Their immune activation mechanisms, including upregulating the proinflammatory cytokines, serving as tumor vaccines, inhibiting immune checkpoints, and modulating intratumoral immune cells, are elaborated. The advantages and disadvantages, as well as possible directions for their clinical translation, are summarized at last.
Collapse
Affiliation(s)
- Wenlu Yan
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Cao
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
23
|
Song B, Wang X, Qin L, Hussain S, Liang W. Brain gliomas: Diagnostic and therapeutic issues and the prospects of drug-targeted nano-delivery technology. Pharmacol Res 2024; 206:107308. [PMID: 39019336 DOI: 10.1016/j.phrs.2024.107308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Glioma is the most common intracranial malignant tumor, with severe difficulty in treatment and a low patient survival rate. Due to the heterogeneity and invasiveness of tumors, lack of personalized clinical treatment design, and physiological barriers, it is often difficult to accurately distinguish gliomas, which dramatically affects the subsequent diagnosis, imaging treatment, and prognosis. Fortunately, nano-delivery systems have demonstrated unprecedented capabilities in diagnosing and treating gliomas in recent years. They have been modified and surface modified to efficiently traverse BBB/BBTB, target lesion sites, and intelligently release therapeutic or contrast agents, thereby achieving precise imaging and treatment. In this review, we focus on nano-delivery systems. Firstly, we provide an overview of the standard and emerging diagnostic and treatment technologies for glioma in clinical practice. After induction and analysis, we focus on summarizing the delivery methods of drug delivery systems, the design of nanoparticles, and their new advances in glioma imaging and treatment in recent years. Finally, we discussed the prospects and potential challenges of drug-delivery systems in diagnosing and treating glioma.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Shehbaz Hussain
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
24
|
Xu C, Jiang C, Li Z, Gao H, Xian J, Guo W, He D, Peng X, Zhou D, Li D. Exosome nanovesicles: biomarkers and new strategies for treatment of human diseases. MedComm (Beijing) 2024; 5:e660. [PMID: 39015555 PMCID: PMC11247338 DOI: 10.1002/mco2.660] [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: 02/21/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024] Open
Abstract
Exosomes are nanoscale vesicles of cellular origin. One of the main characteristics of exosomes is their ability to carry a wide range of biomolecules from their parental cells, which are important mediators of intercellular communication and play an important role in physiological and pathological processes. Exosomes have the advantages of biocompatibility, low immunogenicity, and wide biodistribution. As researchers' understanding of exosomes has increased, various strategies have been proposed for their use in diagnosing and treating diseases. Here, we provide an overview of the biogenesis and composition of exosomes, describe the relationship between exosomes and disease progression, and focus on the use of exosomes as biomarkers for early screening, disease monitoring, and guiding therapy in refractory diseases such as tumors and neurodegenerative diseases. We also summarize the current applications of exosomes, especially engineered exosomes, for efficient drug delivery, targeted therapies, gene therapies, and immune vaccines. Finally, the current challenges and potential research directions for the clinical application of exosomes are also discussed. In conclusion, exosomes, as an emerging molecule that can be used in the diagnosis and treatment of diseases, combined with multidisciplinary innovative solutions, will play an important role in clinical applications.
Collapse
Affiliation(s)
- Chuan Xu
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Chaoyang Jiang
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Zhihui Li
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Hui Gao
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Jing Xian
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Wenyan Guo
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Dan He
- Department of OncologyThe Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanChina
| | - Xingchen Peng
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Daijun Zhou
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| | - Dong Li
- Department of OncologyThe General Hospital of Western Theater CommandChengduChina
| |
Collapse
|
25
|
Susa F, Arpicco S, Pirri CF, Limongi T. An Overview on the Physiopathology of the Blood-Brain Barrier and the Lipid-Based Nanocarriers for Central Nervous System Delivery. Pharmaceutics 2024; 16:849. [PMID: 39065547 PMCID: PMC11279990 DOI: 10.3390/pharmaceutics16070849] [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: 05/11/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The state of well-being and health of our body is regulated by the fine osmotic and biochemical balance established between the cells of the different tissues, organs, and systems. Specific districts of the human body are defined, kept in the correct state of functioning, and, therefore, protected from exogenous or endogenous insults of both mechanical, physical, and biological nature by the presence of different barrier systems. In addition to the placental barrier, which even acts as a linker between two different organisms, the mother and the fetus, all human body barriers, including the blood-brain barrier (BBB), blood-retinal barrier, blood-nerve barrier, blood-lymph barrier, and blood-cerebrospinal fluid barrier, operate to maintain the physiological homeostasis within tissues and organs. From a pharmaceutical point of view, the most challenging is undoubtedly the BBB, since its presence notably complicates the treatment of brain disorders. BBB action can impair the delivery of chemical drugs and biopharmaceuticals into the brain, reducing their therapeutic efficacy and/or increasing their unwanted bioaccumulation in the surrounding healthy tissues. Recent nanotechnological innovation provides advanced biomaterials and ad hoc customized engineering and functionalization methods able to assist in brain-targeted drug delivery. In this context, lipid nanocarriers, including both synthetic (liposomes, solid lipid nanoparticles, nanoemulsions, nanostructured lipid carriers, niosomes, proniosomes, and cubosomes) and cell-derived ones (extracellular vesicles and cell membrane-derived nanocarriers), are considered one of the most successful brain delivery systems due to their reasonable biocompatibility and ability to cross the BBB. This review aims to provide a complete and up-to-date point of view on the efficacy of the most varied lipid carriers, whether FDA-approved, involved in clinical trials, or used in in vitro or in vivo studies, for the treatment of inflammatory, cancerous, or infectious brain diseases.
Collapse
Affiliation(s)
- Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| | - Candido Fabrizio Pirri
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Tania Limongi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| |
Collapse
|
26
|
Cela I, Capone E, Trevisi G, Sala G. Extracellular vesicles in glioblastoma: Biomarkers and therapeutic tools. Semin Cancer Biol 2024; 101:25-43. [PMID: 38754752 DOI: 10.1016/j.semcancer.2024.04.003] [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: 12/01/2023] [Revised: 03/19/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
Glioblastoma (GBM) is the most aggressive tumor among the gliomas and intracranial tumors and to date prognosis for GBM patients remains poor, with a median survival typically measured in months to a few years depending on various factors. Although standardized therapies are routinely employed, it is clear that these strategies are unable to cope with heterogeneity and invasiveness of GBM. Furthermore, diagnosis and monitoring of responses to therapies are directly dependent on tissue biopsies or magnetic resonance imaging (MRI) techniques. From this point of view, liquid biopsies are arising as key sources of a variety of biomarkers with the advantage of being easily accessible and monitorable. In this context, extracellular vesicles (EVs), physiologically shed into body fluids by virtually all cells, are gaining increasing interest both as natural carriers of biomarkers and as specific signatures even for GBM. What makes these vesicles particularly attractive is they are also emerging as therapeutical vehicles to treat GBM given their native ability to cross the blood-brain barrier (BBB). Here, we reviewed recent advances on the use of EVs as biomarker for liquid biopsy and nanocarriers for targeted delivery of anticancer drugs in glioblastoma.
Collapse
Affiliation(s)
- Ilaria Cela
- Department of Innovative Technologies in Medicine & Dentistry, University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Emily Capone
- Department of Innovative Technologies in Medicine & Dentistry, University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Gianluca Trevisi
- Department of Neurosciences, Imaging and Clinical Sciences, "G. D'Annunzio" University, Chieti, Italy; Neurosurgical Unit, Santo Spirito Hospital, Pescara 65121, Italy
| | - Gianluca Sala
- Department of Innovative Technologies in Medicine & Dentistry, University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
27
|
Liang R, Lu H, Zhu H, Liang G, Zhang J, Gao J, Tian T. Radiation-primed TGF-β trapping by engineered extracellular vesicles for targeted glioblastoma therapy. J Control Release 2024; 370:821-834. [PMID: 38740092 DOI: 10.1016/j.jconrel.2024.05.022] [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: 02/05/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
The poor outcome of glioblastoma multiforme (GBM) treated with immunotherapy is attributed to the profound immunosuppressive tumor microenvironment (TME) and the lack of effective delivery across the blood-brain barrier. Radiation therapy (RT) induces an immunogenic antitumor response that is counteracted by evasive mechanisms, among which transforming growth factor-β (TGF-β) activation is the most prominent factor. We report an extracellular vesicle (EV)-based nanotherapeutic that traps TGF-β by expressing the extracellular domain of the TGF-β type II receptor and targets GBM by decorating the EV surface with RGD peptide. We show that short-burst radiation dramatically enhanced the targeting efficiency of RGD peptide-conjugated EVs to GBM, while the displayed TGF-β trap reversed radiation-stimulated TGF-β activation in the TME, offering a synergistic effect in the murine GBM model. The combined therapy significantly increased CD8+ cytotoxic T cells infiltration and M1/M2 macrophage ratio, resulting in the regression of tumor growth and prolongation of overall survival. These results provide an EV-based therapeutic strategy for immune remodeling of the GBM TME and eradication of therapy-resistant tumors, further supporting its clinical translation.
Collapse
Affiliation(s)
- Ruyu Liang
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Hongyu Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China; Department of Neurosurgery, Funing People's Hospital, Funing 224400, Jiangsu, China
| | - Haifeng Zhu
- Department of Neurosurgery, Funing People's Hospital, Funing 224400, Jiangsu, China
| | - Gaofeng Liang
- School of Basic Medicineand Forensic Medicine, Henan University of Science & Technology, Luoyang 471023, Henan, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Jun Gao
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, China.
| | - Tian Tian
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
28
|
Liao C, Huang Z, Liu J, Deng M, Wang L, Chen Y, Li J, Zhao J, Luo X, Zhu J, Wu Q, Fu W, Sun B, Zheng J. Role of extracellular vesicles in castration-resistant prostate cancer. Crit Rev Oncol Hematol 2024; 197:104348. [PMID: 38588967 DOI: 10.1016/j.critrevonc.2024.104348] [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: 03/13/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024] Open
Abstract
Prostate cancer (PCa) is a common health threat to men worldwide, and castration-resistant PCa (CRPC) is the leading cause of PCa-related deaths. Extracellular vesicles (EVs) are lipid bilayer compartments secreted by living cells that are important mediators of intercellular communication. EVs regulate the biological processes of recipient cells by transmitting heterogeneous cargoes, contributing to CRPC occurrence, progression, and drug resistance. These EVs originate not only from malignant cells, but also from various cell types within the tumor microenvironment. EVs are widely dispersed throughout diverse biological fluids and are attractive biomarkers derived from noninvasive liquid biopsy techniques. EV quantities and cargoes have been tested as potential biomarkers for CRPC diagnosis, progression, drug resistance, and prognosis; however, technical barriers to their clinical application continue to exist. Furthermore, exogenous EVs may provide tools for new therapies for CRPC. This review summarizes the current evidence on the role of EVs in CRPC.
Collapse
Affiliation(s)
- Chaoyu Liao
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Zeyu Huang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jingui Liu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Min Deng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Leyi Wang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yutong Chen
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jia Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jiang Zhao
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xing Luo
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jingzhen Zhu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Qingjian Wu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Weihua Fu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Bishao Sun
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| |
Collapse
|
29
|
Choi W, Park DJ, Eliceiri BP. Defining tropism and activity of natural and engineered extracellular vesicles. Front Immunol 2024; 15:1363185. [PMID: 38660297 PMCID: PMC11039936 DOI: 10.3389/fimmu.2024.1363185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Extracellular vesicles (EVs) have important roles as mediators of cell-to-cell communication, with physiological functions demonstrated in various in vivo models. Despite advances in our understanding of the biological function of EVs and their potential for use as therapeutics, there are limitations to the clinical approaches for which EVs would be effective. A primary determinant of the biodistribution of EVs is the profile of proteins and other factors on the surface of EVs that define the tropism of EVs in vivo. For example, proteins displayed on the surface of EVs can vary in composition by cell source of the EVs and the microenvironment into which EVs are delivered. In addition, interactions between EVs and recipient cells that determine uptake and endosomal escape in recipient cells affect overall systemic biodistribution. In this review, we discuss the contribution of the EV donor cell and the role of the microenvironment in determining EV tropism and thereby determining the uptake and biological activity of EVs.
Collapse
Affiliation(s)
- Wooil Choi
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Dong Jun Park
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Brian P. Eliceiri
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
- Department of Dermatology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
30
|
Cao J, Lv G, Wei F. Engineering exosomes to reshape the immune microenvironment in breast cancer: Molecular insights and therapeutic opportunities. Clin Transl Med 2024; 14:e1645. [PMID: 38572668 PMCID: PMC10993163 DOI: 10.1002/ctm2.1645] [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: 11/28/2023] [Revised: 02/19/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Breast cancer remains a global health challenge, necessitating innovative therapeutic approaches. Immunomodulation and immunotherapy have emerged as promising strategies for breast cancer treatment. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. Through suitable modifications, engineered exosomes exhibit the capability to overcome the limitations associated with traditional therapeutic approaches. This ability opens up novel avenues for the development of more effective, personalized, and minimally invasive interventions. MAIN BODY In this comprehensive review, we explore the molecular insights and therapeutic potential of engineered exosomes in breast cancer. We discuss the strategies employed for exosome engineering and delve into their molecular mechanisms in reshaping the immune microenvironment of breast cancer. CONCLUSIONS By elucidating the contribution of engineered exosomes to breast cancer immunomodulation, this review underscores the transformative potential of this emerging field for improving breast cancer therapy. HIGHLIGHTS Surface modification of exosomes can improve the targeting specificity. The engineered exosome-loaded immunomodulatory cargo regulates the tumour immune microenvironment. Engineered exosomes are involved in the immune regulation of breast cancer.
Collapse
Affiliation(s)
- Jilong Cao
- Party Affairs and Administration Officethe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| | - Gang Lv
- Department of Thyroid and Breast SurgeryChaohu Hospital of Anhui Medical UniversityChaohuP. R. China
| | - Fang Wei
- Department of General Surgerythe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| |
Collapse
|
31
|
Hsia T, Chen Y. RNA-encapsulating lipid nanoparticles in cancer immunotherapy: From pre-clinical studies to clinical trials. Eur J Pharm Biopharm 2024; 197:114234. [PMID: 38401743 DOI: 10.1016/j.ejpb.2024.114234] [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: 11/19/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2024]
Abstract
Nanoparticle-based delivery systems such as RNA-encapsulating lipid nanoparticles (RNA LNPs) have dramatically advanced in function and capacity over the last few decades. RNA LNPs boast of a diverse array of external and core configurations that enhance targeted delivery and prolong circulatory retention, advancing therapeutic outcomes. Particularly within the realm of cancer immunotherapies, RNA LNPs are increasingly gaining prominence. Pre-clinical in vitro and in vivo studies have laid a robust foundation for new and ongoing clinical trials that are actively enrolling patients for RNA LNP cancer immunotherapy. This review explores RNA LNPs, starting from their core composition to their external membrane formulation, set against a backdrop of recent clinical breakthroughs. We further elucidate the LNP delivery avenues, broach the prevailing challenges, and contemplate the future perspectives of RNA LNP-mediated immunotherapy.
Collapse
Affiliation(s)
- Tiffaney Hsia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
32
|
Si C, Gao J, Ma X. Engineered exosomes in emerging cell-free therapy. Front Oncol 2024; 14:1382398. [PMID: 38595822 PMCID: PMC11003191 DOI: 10.3389/fonc.2024.1382398] [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: 02/05/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
The discovery and use of exosomes ushered in a new era of cell-free therapy. Exosomes are a subgroup of extracellular vesicles that show great potential in disease treatment. Engineered exosomes. with their improved functions have attracted intense interests of their application in translational medicine research. However, the technology of engineering exosomes still faces many challenges which have been the great limitation for their clinical application. This review summarizes the current status of research on engineered exosomes and the difficulties encountered in recent years, with a view to providing new approaches and ideas for future exosome modification and new drug development.
Collapse
Affiliation(s)
| | - Jianen Gao
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xu Ma
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Schweiger MW, Amoozgar Z, Repiton P, Morris R, Maksoud S, Hla M, Zaniewski E, Noske DP, Haas W, Breyne K, Tannous BA. Glioblastoma extracellular vesicles modulate immune PD-L1 expression in accessory macrophages upon radiotherapy. iScience 2024; 27:108807. [PMID: 38303726 PMCID: PMC10831876 DOI: 10.1016/j.isci.2024.108807] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/10/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, presenting major challenges due to limited treatment options. Standard care includes radiation therapy (RT) to curb tumor growth and alleviate symptoms, but its impact on GBM is limited. In this study, we investigated the effect of RT on immune suppression and whether extracellular vesicles (EVs) originating from GBM and taken up by the tumor microenvironment (TME) contribute to the induced therapeutic resistance. We observed that (1) ionizing radiation increases immune-suppressive markers on GBM cells, (2) macrophages exacerbate immune suppression in the TME by increasing PD-L1 in response to EVs derived from GBM cells which is further modulated by RT, and (3) RT increases CD206-positive macrophages which have the most potential in inducing a pro-oncogenic environment due to their increased uptake of tumor-derived EVs. In conclusion, RT affects GBM resistance by immuno-modulating EVs taken up by myeloid cells in the TME.
Collapse
Affiliation(s)
- Markus W. Schweiger
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Neurosurgery, 1081 HV Amsterdam, the Netherlands
- Cancer Center Amsterdam, Brain Tumor Center and Liquid Biopsy Center, 1081 HV Amsterdam, the Netherlands
| | - Zohreh Amoozgar
- Department of Radiation Oncology, Edwin L. Steele Laboratories, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pierre Repiton
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1205 Geneva, Switzerland
| | - Robert Morris
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA
| | - Semer Maksoud
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Michael Hla
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Eric Zaniewski
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA
| | - David P. Noske
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Neurosurgery, 1081 HV Amsterdam, the Netherlands
- Cancer Center Amsterdam, Brain Tumor Center and Liquid Biopsy Center, 1081 HV Amsterdam, the Netherlands
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Boston, MA 02129, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02129, USA
| | - Koen Breyne
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Bakhos A. Tannous
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
34
|
Onkar A, Khan F, Goenka A, Rajendran RL, Dmello C, Hong CM, Mubin N, Gangadaran P, Ahn BC. Smart Nanoscale Extracellular Vesicles in the Brain: Unveiling their Biology, Diagnostic Potential, and Therapeutic Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6709-6742. [PMID: 38315446 DOI: 10.1021/acsami.3c16839] [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: 02/07/2024]
Abstract
Information exchange is essential for the brain, where it communicates the physiological and pathological signals to the periphery and vice versa. Extracellular vesicles (EVs) are a heterogeneous group of membrane-bound cellular informants actively transferring informative calls to and from the brain via lipids, proteins, and nucleic acid cargos. In recent years, EVs have also been widely used to understand brain function, given their "cell-like" properties. On the one hand, the presence of neuron and astrocyte-derived EVs in biological fluids have been exploited as biomarkers to understand the mechanisms and progression of multiple neurological disorders; on the other, EVs have been used in designing targeted therapies due to their potential to cross the blood-brain-barrier (BBB). Despite the expanding literature on EVs in the context of central nervous system (CNS) physiology and related disorders, a comprehensive compilation of the existing knowledge still needs to be made available. In the current review, we provide a detailed insight into the multifaceted role of brain-derived extracellular vesicles (BDEVs) in the intricate regulation of brain physiology. Our focus extends to the significance of these EVs in a spectrum of disorders, including brain tumors, neurodegenerative conditions, neuropsychiatric diseases, autoimmune disorders, and others. Throughout the review, parallels are drawn for using EVs as biomarkers for various disorders, evaluating their utility in early detection and monitoring. Additionally, we discuss the promising prospects of utilizing EVs in targeted therapy while acknowledging the existing limitations and challenges associated with their applications in clinical scenarios. A foundational comprehension of the current state-of-the-art in EV research is essential for informing the design of future studies.
Collapse
Affiliation(s)
- Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California 94143, United States
| | - Fatima Khan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Crismita Dmello
- Department of Neurological Surgery and Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Nida Mubin
- Department of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
35
|
Das A, Fernandez NR, Levine A, Bianchi V, Stengs LK, Chung J, Negm L, Dimayacyac JR, Chang Y, Nobre L, Ercan AB, Sanchez-Ramirez S, Sudhaman S, Edwards M, Larouche V, Samuel D, Van Damme A, Gass D, Ziegler DS, Bielack SS, Koschmann C, Zelcer S, Yalon-Oren M, Campino GA, Sarosiek T, Nichols KE, Loret De Mola R, Bielamowicz K, Sabel M, Frojd CA, Wood MD, Glover JM, Lee YY, Vanan M, Adamski JK, Perreault S, Chamdine O, Hjort MA, Zapotocky M, Carceller F, Wright E, Fedorakova I, Lossos A, Tanaka R, Osborn M, Blumenthal DT, Aronson M, Bartels U, Huang A, Ramaswamy V, Malkin D, Shlien A, Villani A, Dirks PB, Pugh TJ, Getz G, Maruvka YE, Tsang DS, Ertl-Wagner B, Hawkins C, Bouffet E, Morgenstern DA, Tabori U. Combined Immunotherapy Improves Outcome for Replication-Repair-Deficient (RRD) High-Grade Glioma Failing Anti-PD-1 Monotherapy: A Report from the International RRD Consortium. Cancer Discov 2024; 14:258-273. [PMID: 37823831 PMCID: PMC10850948 DOI: 10.1158/2159-8290.cd-23-0559] [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: 05/15/2023] [Revised: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 10/13/2023]
Abstract
Immune checkpoint inhibition (ICI) is effective for replication-repair-deficient, high-grade gliomas (RRD-HGG). The clinical/biological impact of immune-directed approaches after failing ICI monotherapy is unknown. We performed an international study on 75 patients treated with anti-PD-1; 20 are progression free (median follow-up, 3.7 years). After second progression/recurrence (n = 55), continuing ICI-based salvage prolonged survival to 11.6 months (n = 38; P < 0.001), particularly for those with extreme mutation burden (P = 0.03). Delayed, sustained responses were observed, associated with changes in mutational spectra and the immune microenvironment. Response to reirradiation was explained by an absence of deleterious postradiation indel signatures (ID8). CTLA4 expression increased over time, and subsequent CTLA4 inhibition resulted in response/stable disease in 75%. RAS-MAPK-pathway inhibition led to the reinvigoration of peripheral immune and radiologic responses. Local (flare) and systemic immune adverse events were frequent (biallelic mismatch-repair deficiency > Lynch syndrome). We provide a mechanistic rationale for the sustained benefit in RRD-HGG from immune-directed/synergistic salvage therapies. Future approaches need to be tailored to patient and tumor biology. SIGNIFICANCE Hypermutant RRD-HGG are susceptible to checkpoint inhibitors beyond initial progression, leading to improved survival when reirradiation and synergistic immune/targeted agents are added. This is driven by their unique biological and immune properties, which evolve over time. Future research should focus on combinatorial regimens that increase patient survival while limiting immune toxicity. This article is featured in Selected Articles from This Issue, p. 201.
Collapse
Affiliation(s)
- Anirban Das
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatric Haematology and Oncology, Tata Medical Center, Kolkata, India
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Nicholas R. Fernandez
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Adrian Levine
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Vanessa Bianchi
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Lucie K. Stengs
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Jiil Chung
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Logine Negm
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Jose Rafael Dimayacyac
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Yuan Chang
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Liana Nobre
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Ayse B. Ercan
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Santiago Sanchez-Ramirez
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Sumedha Sudhaman
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Melissa Edwards
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Valerie Larouche
- Pediatric Haematology/Oncology Department, CHU de Québec-Université Laval, Quebec City, Canada
| | - David Samuel
- Department of Paediatric Oncology, Valley Children's Hospital, Madera, California
| | - An Van Damme
- Department of Paediatric Haematology and Oncology, Saint Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - David Gass
- Atrium Health/Levine Children's Hospital, Charlotte, North Carolina
| | - David S. Ziegler
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
- School of Clinical Medicine, UNSW Sydney, Sydney, Australia
| | - Stefan S. Bielack
- Department of Pediatric Oncology, Hematology and Immunology, Center for Childhood, Adolescent, and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart, Stuttgart, Germany
| | - Carl Koschmann
- Pediatric Hematology/Oncology, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan
| | - Shayna Zelcer
- Department of Pediatrics, London Health Sciences Centre, London, Canada
| | - Michal Yalon-Oren
- Department of Paediatric Haematology-Oncology, Sheba Medical Centre, Ramat Gan, Israel
| | - Gadi Abede Campino
- Department of Paediatric Haematology-Oncology, Sheba Medical Centre, Ramat Gan, Israel
| | | | - Kim E. Nichols
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Kevin Bielamowicz
- Department of Pediatrics, Section of Pediatric Hematology/Oncology, The University of Arkansas for Medical Sciences/Arkansas Children's Hospital, Little Rock, Arkansas
| | - Magnus Sabel
- Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg & Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Charlotta A. Frojd
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Matthew D. Wood
- Neuropathology, Oregon Health & Science University Department of Pathology, Portland, Oregon
| | - Jason M. Glover
- Department of Pediatric Hematology/Oncology, Randall Children's Hospital, Portland, Oregon
| | - Yi-Yen Lee
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Magimairajan Vanan
- Pediatric Hematology-Oncology, CancerCare Manitoba, Winnipeg, Canada
- CancerCare Manitoba Research Institute, Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - Jenny K. Adamski
- Neuro-oncology Division, Birmingham Children's Hospital, Birmingham, United Kingdom
| | - Sebastien Perreault
- Neurosciences Department, Child Neurology Division, CHU Sainte-Justine, Montreal, Canada
| | - Omar Chamdine
- Pediatric Hematology Oncology, King Fahad Specialist Hospital Dammam, Eastern Province, Saudi Arabia
| | - Magnus Aasved Hjort
- Department of Paediatric Haematology and Oncology, St. Olav's University Hospital, Trondheim, Norway
| | - Michal Zapotocky
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Fernando Carceller
- Paediatric and Adolescent Neuro-Oncology and Drug Development, The Royal Marsden NHS Foundation Trust & Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Erin Wright
- Division of Neuro-Oncology, Akron Children's Hospital, Akron, Ohio
| | - Ivana Fedorakova
- Clinic of Pediatric Oncology and Hematology, University Children's Hospital, Banská Bystrica, Slovakia
| | - Alexander Lossos
- Department of Oncology, Leslie and Michael Gaffin Centre for Neuro-Oncology, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Ryuma Tanaka
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael Osborn
- Women's and Children's Hospital, North Adelaide, Australia
| | - Deborah T. Blumenthal
- Neuro-Oncology Service, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Melyssa Aronson
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Canada
| | - Ute Bartels
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Annie Huang
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Vijay Ramaswamy
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - David Malkin
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Adam Shlien
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Anita Villani
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Peter B. Dirks
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Trevor J. Pugh
- Ontario Institute for Cancer Research, Princess Margaret Cancer Centre, Toronto, Canada
| | - Gad Getz
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | - Derek S. Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Birgit Ertl-Wagner
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Eric Bouffet
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Daniel A. Morgenstern
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Uri Tabori
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
36
|
You Q, Liang F, Wu G, Cao F, Liu J, He Z, Wang C, Zhu L, Chen X, Yang Y. The Landscape of Biomimetic Nanovesicles in Brain Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306583. [PMID: 37713652 DOI: 10.1002/adma.202306583] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Brain diseases, such as brain tumors, neurodegenerative diseases, cerebrovascular diseases, and brain injuries, are caused by various pathophysiological changes, which pose a serious health threat. Brain disorders are often difficult to treat due to the presence of the blood-brain barrier (BBB). Biomimetic nanovesicles (BNVs), including endogenous extracellular vesicles (EVs) derived from various cells and artificial nanovesicles, possess the ability to penetrate the BBB and thus can be utilized for drug delivery to the brain. BNVs, especially endogenous EVs, are widely distributed in body fluids and usually carry various disease-related signal molecules such as proteins, RNA, and DNA, and may also be analyzed to understand the etiology and pathogenesis of brain diseases. This review covers the exhaustive classification and characterization of BNVs and pathophysiological roles involved in various brain diseases, and emphatically focuses on nanotechnology-integrated BNVs for brain disease theranostics, including various diagnosis strategies and precise therapeutic regulations (e.g., immunity regulation, disordered protein clearance, anti-neuroinflammation, neuroregeneration, angiogenesis, and the gut-brain axis regulation). The remaining challenges and future perspectives regarding the nanotechnology-integrated BNVs for the diagnosis and treatment of brain diseases are also discussed and outlined.
Collapse
Affiliation(s)
- Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Gege Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Jingyi Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing, 400016, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
37
|
Li X, Wei Y, Zhang Z, Zhang X. Harnessing genetically engineered cell membrane-derived vesicles as biotherapeutics. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:44-63. [PMID: 39698409 PMCID: PMC11648408 DOI: 10.20517/evcna.2023.58] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/22/2023] [Accepted: 01/22/2024] [Indexed: 12/20/2024]
Abstract
Cell membrane-derived vesicles (CMVs) are particles generated from living cells, including extracellular vesicles (EVs) and artificial extracellular vesicles (aEVs) prepared from cell membranes. CMVs possess considerable potential in drug delivery, regenerative medicine, immunomodulation, disease diagnosis, etc. owing to their stable lipid bilayer structure, favorable biocompatibility, and low toxicity. Although the majority of CMVs inherit certain attributes from the original cells, it is still difficult to execute distinct therapeutic functions, such as organ targeting, signal regulation, and exogenous biotherapeutic supplementation. Hence, engineering CMVs by genetic engineering, chemical modification, and hybridization is a promising way to endow CMVs with specific functions and open up novel vistas for applications. In particular, there is a growing interest in genetically engineered CMVs harnessed to exhibit biotherapeutics. Herein, we outline the preparation strategies and their characteristics for purifying CMVs. Additionally, we review the advances of genetically engineered CMVs utilized to target organs, regulate signal transduction, and deliver biomacromolecules and chemical drugs. Furthermore, we also summarize the emerging therapeutic applications of genetically engineered CMVs in addressing tumors, diabetes, systemic lupus erythematosus, and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaohong Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Yuting Wei
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Zhirang Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Xudong Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| |
Collapse
|
38
|
Ahmadi M, Abbasi R, Rezaie J. Tumor immune escape: extracellular vesicles roles and therapeutics application. Cell Commun Signal 2024; 22:9. [PMID: 38167133 PMCID: PMC10763406 DOI: 10.1186/s12964-023-01370-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/28/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Immune escape, a process by which tumor cells evade immune surveillance, remains a challenge for cancer therapy. Tumor cells produce extracellular vesicles (EVs) that participate in immune escape by transferring bioactive molecules between cells. EVs refer to heterogeneous vesicles that participate in intercellular communication. EVs from tumor cells usually carry tumor antigens and have been considered a source of tumor antigens to induce anti-tumor immunity. However, evidence also suggests that these EVs can accelerate immune escape by carrying heat shock proteins (HSPs), programmed death-ligand 1 (PD-L1), etc. to immune cells, suppressing function and exhausting the immune cells pool. EVs are progressively being evaluated for therapeutic implementation in cancer therapies. EVs-based immunotherapies involve inhibiting EVs generation, using natural EVs, and harnessing engineering EVs. All approaches are associated with advantages and disadvantages. The EVs heterogeneity and diverse physicochemical properties are the main challenges to their clinical applications. SHORT CONCLUSION Although EVs are criminal; they can be useful for overcoming immune escape. This review discusses the latest knowledge on EVs population and sheds light on the function of tumor-derived EVs in immune escape. It also describes EVs-based immunotherapies with a focus on engineered EVs, followed by challenges that hinder the clinical translation of EVs that are essential to be addressed in future investigations. Video Abstract.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Abbasi
- Department of Biology, Urmia University, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
39
|
Fei X, Wu J, Tian H, Jiang D, Chen H, Yan K, Wang Y, Zhao Y, Chen H, Xie X, Wang Z, Zhu W, Huang Q. Glioma stem cells remodel immunotolerant microenvironment in GBM and are associated with therapeutic advancements. Cancer Biomark 2024; 41:1-24. [PMID: 39240627 PMCID: PMC11492047 DOI: 10.3233/cbm-230486] [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: 11/15/2023] [Accepted: 07/19/2024] [Indexed: 09/07/2024]
Abstract
Glioma is the most common primary tumor of the central nervous system (CNS). Glioblastoma (GBM) is incurable with current treatment strategies. Additionally, the treatment of recurrent GBM (rGBM) is often referred to as terminal treatment, necessitating hospice-level care and management. The presence of the blood-brain barrier (BBB) gives GBM a more challenging or "cold" tumor microenvironment (TME) than that of other cancers and gloma stem cells (GSCs) play an important role in the TME remodeling, occurrence, development and recurrence of giloma. In this review, our primary focus will be on discussing the following topics: niche-associated GSCs and macrophages, new theories regarding GSC and TME involving pyroptosis and ferroptosis in GBM, metabolic adaptations of GSCs, the influence of the cold environment in GBM on immunotherapy, potential strategies to transform the cold GBM TME into a hot one, and the advancement of GBM immunotherapy and GBM models.
Collapse
Affiliation(s)
- Xifeng Fei
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Jie Wu
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
- Department of Neurosurgery, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing University Medical School, Suzhou, Jiangsu, China
| | - Haiyan Tian
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
- Department of GCP, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Dongyi Jiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Hanchun Chen
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Ke Yan
- Department of Neurosurgery, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing University Medical School, Suzhou, Jiangsu, China
| | - Yuan Wang
- Pediatric Cancer Center, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yaodong Zhao
- Department of Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Hua Chen
- Department of Neurosurgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiangtong Xie
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Zhimin Wang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
- Department of Neurosurgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Wenyu Zhu
- Department of Neurosurgery, The Affiliated Suzhou Science and Technology Town Hospital of Nanjing University Medical School, Suzhou, Jiangsu, China
| | - Qiang Huang
- Department of Neurosurgery, Second Affiliated Hospital of Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
40
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
41
|
Liu H, Luo GF, Shang Z. Plant-derived nanovesicles as an emerging platform for cancer therapy. Acta Pharm Sin B 2024; 14:133-154. [PMID: 38239235 PMCID: PMC10792991 DOI: 10.1016/j.apsb.2023.08.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/14/2023] [Accepted: 08/26/2023] [Indexed: 01/22/2024] Open
Abstract
Plant-derived nanovesicles (PDNVs) derived from natural green products have emerged as an attractive nanoplatform in biomedical application. They are usually characterized by unique structural and biological functions, such as the bioactive lipids/proteins/nucleic acids as therapeutics and targeting groups, immune-modulation, and long-term circulation. With the rapid development of nanotechnology, materials, and synthetic chemistry, PDNVs can be engineered with multiple functions for efficient drug delivery and specific killing of diseased cells, which represent an innovative biomaterial with high biocompatibility for fighting against cancer. In this review, we provide an overview of the state-of-the-art studies concerning the development of PDNVs for cancer therapy. The original sources, methods for obtaining PDNVs, composition and structure are introduced systematically. With an emphasis on the featured application, the inherent anticancer properties of PDNVs as well as the strategies in constructing multifunctional PDNVs-based nanomaterials will be discussed in detail. Finally, some scientific issues and technical challenges of PDNVs as promising options in improving anticancer therapy will be discussed, which are expected to promote the further development of PDNVs in clinical translation.
Collapse
Affiliation(s)
- Hanzhe Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Guo-Feng Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhengjun Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
42
|
Wang W, Zhang Y, Jian Y, He S, Liu J, Cheng Y, Zheng S, Qian Z, Gao X, Wang X. Sensitizing chemotherapy for glioma with fisetin mediated by a microenvironment-responsive nano-drug delivery system. NANOSCALE 2023; 16:97-109. [PMID: 38087978 DOI: 10.1039/d3nr05195a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Drug resistance has become an obstacle to successful cancer chemotherapies, with therapeutic agents effectively traversing the blood-brain barrier (BBB) remaining a great challenge. A microenvironment responsive and active targeting nanoparticle was constructed to enhance the penetration of drugs, leading to improved therapeutic effects. Dynamic light scattering demonstrated that the prepared nanoparticle had a uniform size. The cRGD modification renders the nanoparticle with active targeting capabilities to traverse the BBB for chemotherapy. The disulfide-bond-containing nanoparticle can be disintegrated in response to a high concentration of endogenous glutathione (GSH) within the tumor microenvironment (TME) for tumor-specific drug release, resulting in more effective accumulation. Notably, the released fisetin further increased the uptake of doxorubicin by glioma cells and exerted synergistic effects to promote apoptosis, induce cellular G2/M cycle arrest, and inhibit cell proliferation and migration in vitro. Moreover, the nanoparticle showed favorable anti-glioma effects in vivo. Our study provides a new strategy to overcome drug resistance by utilizing a natural product to sensitize conventional chemotherapeutics with well-designed targeted nanodelivery systems for cancer treatment.
Collapse
Affiliation(s)
- Wanyu Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Yuanyuan Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yue Jian
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Shi He
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Jiagang Liu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Yongzhong Cheng
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Songping Zheng
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Zhiyong Qian
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Xiang Gao
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Xiang Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
43
|
Wei YN, Yan CY, Zhao ML, Zhao XH. The role and application of vesicles in triple-negative breast cancer: Opportunities and challenges. Mol Ther Oncolytics 2023; 31:100752. [PMID: 38130701 PMCID: PMC10733704 DOI: 10.1016/j.omto.2023.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Extracellular vesicles (EVs) carry DNA, RNA, protein, and other substances involved in intercellular crosstalk and can be used for the targeted delivery of drugs. Triple-negative breast cancer (TNBC) is rich in recurrent and metastatic disease and lacks therapeutic targets. Studies have proved the role of EVs in the different stages of the genesis and development of TNBC. Cancer cells actively secrete various biomolecules, which play a significant part establishing the tumor microenvironment via EVs. In this article, we describe the roles of EVs in the tumor immune microenvironment, metabolic microenvironment, and vascular remodeling, and summarize the application of EVs for objective delivery of chemotherapeutic drugs, immune antigens, and cancer vaccine adjuvants. EVs-based therapy may represent the next-generation tool for targeted drug delivery for the cure of a variety of diseases lacking effective drug treatment.
Collapse
Affiliation(s)
- Ya-Nan Wei
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Chun-Yan Yan
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Meng-Lu Zhao
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Xi-He Zhao
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| |
Collapse
|
44
|
Shi R, Zhan A, Li X, Kong B, Liang G. Biomimetic extracellular vesicles for the tumor targeted treatment. ENGINEERED REGENERATION 2023; 4:427-437. [DOI: 10.1016/j.engreg.2023.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
45
|
Dong Y, Xia P, Xu X, Shen J, Ding Y, Jiang Y, Wang H, Xie X, Zhang X, Li W, Li Z, Wang J, Zhao SC. Targeted delivery of organic small-molecule photothermal materials with engineered extracellular vesicles for imaging-guided tumor photothermal therapy. J Nanobiotechnology 2023; 21:442. [PMID: 37993888 PMCID: PMC10666357 DOI: 10.1186/s12951-023-02133-5] [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: 07/24/2023] [Accepted: 09/28/2023] [Indexed: 11/24/2023] Open
Abstract
Imaging-guided photothermal therapy (PTT) for cancers recently gathered increasing focus thanks to its precise diagnosis and potent therapeutic effectiveness. Croconaine (CR) dyes demonstrate potential in expanding utility for near infrared (NIR) dyes in bio-imaging/theranostics. However, reports on CR dyes for PTT are scarce most likely due to the short of the efficacious delivery strategies to achieve specific accumulation in diseased tissues to induce PTT. Extracellular vesicles (EVs) are multifunctional nanoparticle systems that function as safe platform for disease theragnostics, which provide potential benefits in extensive biomedical applications. Here, we developed a novel delivery system for photothermal molecules based on a CR dye that exerts photothermal activity through CDH17 nanobody-engineered EVs. The formed CR@E8-EVs showed strong NIR absorption, excellent photothermal performance, good biological compatibility and superb active tumor-targeting capability. The CR@E8-EVs can not only visualize and feature the tumors through CR intrinsic property as a photoacoustic imaging (PAI) agent, but also effectively retard the tumor growth under laser irradiation to perform PTT. It is expected that the engineered EVs will become a novel delivery vehicle of small organic photothermal agents (SOPTAs) in future clinical PTT applications.
Collapse
Affiliation(s)
- Yafang Dong
- Department of Urology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510500, P. R. China
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Peng Xia
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430072, P. R. China
| | - Xiaolong Xu
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Jing Shen
- Department of Oncology, Department of Infectious Disease, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Youbin Ding
- Department of Medical Imaging, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510630, P. R. China
| | - Yuke Jiang
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Huifang Wang
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Xin Xie
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China
| | - Xiaodong Zhang
- Department of Medical Imaging, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510630, P. R. China
| | - Weihua Li
- Medical imaging department, Shenzhen Second People's Hospital/the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, Guangdong, 518035, P. R. China.
| | - Zhijie Li
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China.
| | - Jigang Wang
- Department of Urology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510500, P. R. China.
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, P. R. China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China.
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.
| | - Shan-Chao Zhao
- Department of Urology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510500, P. R. China.
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China.
| |
Collapse
|
46
|
Zhang L, Zhang Y, Wang X, Zhou Y, Qi J, Gu L, Zhao Q, Yu R, Zhou X. A Trojan-Horse-Like Biomimetic Nano-NK to Elicit an Immunostimulatory Tumor Microenvironment for Enhanced GBM Chemo-Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301439. [PMID: 37420326 DOI: 10.1002/smll.202301439] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/20/2023] [Indexed: 07/09/2023]
Abstract
Although the chemo- and immuno-therapies have obtained good responses for several solid tumors, including those with brain metastasis, their clinical efficacy in glioblastoma (GBM) is disappointing. The lack of safe and effective delivery systems across the blood-brain barrier (BBB) and the immunosuppressive tumor microenvironment (TME) are two main hurdles for GBM therapy. Herein, a Trojan-horse-like nanoparticle system is designed, which encapsulates biocompatible PLGA-coated temozolomide (TMZ) and IL-15 nanoparticles (NPs) with cRGD-decorated NK cell membrane (R-NKm@NP), to elicit the immunostimulatory TME for GBM chemo-immunotherapy. Taking advantage of the outer NK cell membrane cooperating with cRGD, the R-NKm@NPs effectively traversed across the BBB and targeted GBM. In addition, the R-NKm@NPs exhibited good antitumor ability and prolonged the median survival of GBM-bearing mice. Notably, after R-NKm@NPs treatment, the locally released TMZ and IL-15 synergistically stimulated the proliferation and activation of NK cells, leading to the maturation of dendritic cells and infiltration of CD8+ cytotoxic T cells, eliciting an immunostimulatory TME. Lastly, the R-NKm@NPs not only effectively prolonged the metabolic cycling time of the drugs in vivo, but also has no noticeable side effects. This study may offer valuable insights for developing biomimetic nanoparticles to potentiate GBM chemo- and immuno-therapies in the future.
Collapse
Affiliation(s)
- Long Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yining Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xu Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yi Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ji Qi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Linbo Gu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Qiu Zhao
- Department of Anesthesiology, The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, Anhui, 230001, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 2210002, China
| |
Collapse
|
47
|
Fadera S, Chukwu C, Stark AH, Yue Y, Xu L, Chien CY, Yuan J, Chen H. Focused Ultrasound-Mediated Delivery of Anti-Programmed Cell Death-Ligand 1 Antibody to the Brain of a Porcine Model. Pharmaceutics 2023; 15:2479. [PMID: 37896238 PMCID: PMC10610297 DOI: 10.3390/pharmaceutics15102479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionized cancer treatment by leveraging the body's immune system to combat cancer cells. However, its effectiveness in brain cancer is hindered by the blood-brain barrier (BBB), impeding the delivery of ICIs to brain tumor cells. This study aimed to assess the safety and feasibility of using focused ultrasound combined with microbubble-mediated BBB opening (FUS-BBBO) to facilitate trans-BBB delivery of an ICI, anti-programmed cell death-ligand 1 antibody (aPD-L1) to the brain of a large animal model. In a porcine model, FUS sonication of targeted brain regions was performed after intravenous microbubble injection, which was followed by intravenous administration of aPD-L1 labeled with a near-infrared fluorescent dye. The permeability of the BBB was evaluated using contrast-enhanced MRI in vivo, while fluorescence imaging and histological analysis were conducted on ex vivo pig brains. Results showed a significant 4.8-fold increase in MRI contrast-enhancement volume in FUS-targeted regions compared to nontargeted regions. FUS sonication enhanced aPD-L1 delivery by an average of 2.1-fold, according to fluorescence imaging. In vivo MRI and ex vivo staining revealed that the procedure did not cause significant acute tissue damage. These findings demonstrate that FUS-BBBO offers a noninvasive, localized, and safe delivery approach for ICI delivery in a large animal model, showcasing its potential for clinical translation.
Collapse
Affiliation(s)
- Siaka Fadera
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
| | - Chinwendu Chukwu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
| | - Andrew H. Stark
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
| | - Lu Xu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
| | - Chih-Yen Chien
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
| | - Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; (S.F.); (Y.Y.); (J.Y.)
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
48
|
Ziegler JN, Tian C. Engineered Extracellular Vesicles: Emerging Therapeutic Strategies for Translational Applications. Int J Mol Sci 2023; 24:15206. [PMID: 37894887 PMCID: PMC10607082 DOI: 10.3390/ijms242015206] [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: 09/25/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Extracellular vesicles (EVs) are small, membrane-bound vesicles used by cells to deliver biological cargo such as proteins, mRNA, and other biomolecules from one cell to another, thus inducing a specific response in the target cell and are a powerful method of cell to cell and organ to organ communication, especially during the pathogenesis of human disease. Thus, EVs may be utilized as prognostic and diagnostic biomarkers, but they also hold therapeutic potential just as mesenchymal stem cells have been used in therapeutics. However, unmodified EVs exhibit poor targeting efficacy, leading to the necessity of engineered EVS. To highlight the advantages and therapeutic promises of engineered EVs, in this review, we summarized the research progress on engineered EVs in the past ten years, especially in the past five years, and highlighted their potential applications in therapeutic development for human diseases. Compared to the existing stem cell-derived EV-based therapeutic strategies, engineered EVs show greater promise in clinical applications: First, engineered EVs mediate good targeting efficacy by exhibiting a targeting peptide that allows them to specifically target a specific organ or even cell type, thus avoiding accumulation in undesired locations and increasing the potency of the treatment. Second, engineered EVs can be artificially pre-loaded with any necessary biomolecular cargo or even therapeutic drugs to treat a variety of human diseases such as cancers, neurological diseases, and cardiovascular ailments. Further research is necessary to improve logistical challenges in large-scale engineered EV manufacturing, but current developments in engineered EVs prove promising to greatly improve therapeutic treatment for traditionally difficult to treat diseases.
Collapse
Affiliation(s)
| | - Changhai Tian
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA;
| |
Collapse
|
49
|
Sharma S, Chepurna O, Sun T. Drug resistance in glioblastoma: from chemo- to immunotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:688-708. [PMID: 38239396 PMCID: PMC10792484 DOI: 10.20517/cdr.2023.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/25/2023] [Indexed: 01/22/2024]
Abstract
As the most common and aggressive type of primary brain tumor in adults, glioblastoma is estimated to end over 10,000 lives each year in the United States alone. Stand treatment for glioblastoma, including surgery followed by radiotherapy and chemotherapy (i.e., Temozolomide), has been largely unchanged since early 2000. Cancer immunotherapy has significantly shifted the paradigm of cancer management in the past decade with various degrees of success in treating many hematopoietic cancers and some solid tumors, such as melanoma and non-small cell lung cancer (NSCLC). However, little progress has been made in the field of neuro-oncology, especially in the application of immunotherapy to glioblastoma treatment. In this review, we attempted to summarize the common drug resistance mechanisms in glioblastoma from Temozolomide to immunotherapy. Our intent is not to repeat the well-known difficulty in the area of neuro-oncology, such as the blood-brain barrier, but to provide some fresh insights into the molecular mechanisms responsible for resistance by summarizing some of the most recent literature. Through this review, we also hope to share some new ideas for improving the immunotherapy outcome of glioblastoma treatment.
Collapse
Affiliation(s)
| | | | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
50
|
Kang J, Mun D, Chun Y, Park D, Kim H, Yun N, Joung B. Engineered small extracellular vesicle-mediated NOX4 siRNA delivery for targeted therapy of cardiac hypertrophy. J Extracell Vesicles 2023; 12:e12371. [PMID: 37795828 PMCID: PMC10552075 DOI: 10.1002/jev2.12371] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/18/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023] Open
Abstract
Small-interfering RNA (siRNA) therapy is considered a powerful therapeutic strategy for treating cardiac hypertrophy, an important risk factor for subsequent cardiac morbidity and mortality. However, the lack of safe and efficient in vivo delivery of siRNAs is a major challenge for broadening its clinical applications. Small extracellular vesicles (sEVs) are a promising delivery system for siRNAs but have limited cell/tissue-specific targeting ability. In this study, a new generation of heart-targeting sEVs (CEVs) has been developed by conjugating cardiac-targeting peptide (CTP) to human peripheral blood-derived sEVs (PB-EVs), using a simple, rapid and scalable method based on bio-orthogonal copper-free click chemistry. The experimental results show that CEVs have typical sEVs properties and excellent heart-targeting ability. Furthermore, to treat cardiac hypertrophy, CEVs are loaded with NADPH Oxidase 4 (NOX4) siRNA (siNOX4). Consequently, CEVs@siNOX4 treatment enhances the in vitro anti-hypertrophic effects by CEVs with siRNA protection and heart-targeting ability. In addition, the intravenous injection of CEVs@siNOX4 into angiotensin II (Ang II)-treated mice significantly improves cardiac function and reduces fibrosis and cardiomyocyte cross-sectional area, with limited side effects. In conclusion, the utilization of CEVs represents an efficient strategy for heart-targeted delivery of therapeutic siRNAs and holds great promise for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Ji‐Young Kang
- Division of Cardiology, Department of Internal MedicineYonsei University College of MedicineSeodaemun‐guSeoulRepublic of Korea
| | - Dasom Mun
- Division of Cardiology, Department of Internal MedicineYonsei University College of MedicineSeodaemun‐guSeoulRepublic of Korea
| | - Yumin Chun
- Division of Cardiology, Department of Internal MedicineYonsei University College of MedicineSeodaemun‐guSeoulRepublic of Korea
| | - Da‐Seul Park
- Division of Cardiology, Department of Internal MedicineYonsei University College of MedicineSeodaemun‐guSeoulRepublic of Korea
| | - Hyoeun Kim
- Department of Biochemistry and Molecular BiologyYonsei University College of MedicineSeodaemun‐guSeoulRepublic of Korea
| | - Nuri Yun
- GNTPharma Science and Technology Center for Health, Giheung‐guYongin‐siIncheonRepublic of Korea
| | - Boyoung Joung
- Division of Cardiology, Department of Internal MedicineYonsei University College of MedicineSeodaemun‐guSeoulRepublic of Korea
- Graduate School of Medical Science, Brain Korea 21 ProjectYonsei University College of MedicineSeodaemun‐guSeoulRepublic of Korea
| |
Collapse
|