1
|
Mao L, Lu J, Wen X, Song Z, Sun C, Zhao Y, Huang F, Chen S, Jiang D, Che W, Zhong C, Yu C, Li K, Lu X, Shi J. Cuproptosis: mechanisms and nanotherapeutic strategies in cancer and beyond. Chem Soc Rev 2025. [PMID: 40433941 DOI: 10.1039/d5cs00083a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Cuproptosis, a novel form of copper (Cu)-dependent programmed cell death, is induced by directly binding Cu species to lipoylated components of the tricarboxylic acid (TCA) cycle. Since its discovery in 2022, cuproptosis has been closely linked to the field of materials science, offering a biological basis and bright prospects for the use of Cu-based nanomaterials in various disease treatments. Owing to the unique physicochemical properties of nanomaterials, Cu delivery nanosystems can specifically increase Cu levels at disease sites, inducing cuproptosis to achieve disease treatment while minimizing the undesirable release of Cu in normal tissues. This innovative nanomaterial-mediated cuproptosis, termed as "nanocuproptosis", positions at the intersection of chemistry, materials science, pharmaceutical science, and clinical medicine. This review aims to comprehensively summarize and discuss recent advancements in cuproptosis across various diseases, with a particular focus on cancer. It delves into the biochemical basis of nanomaterial-mediated cuproptosis, the rational design for cuproptosis inducers, strategies for enhancing therapeutic specificity, and cuproptosis-centric synergistic cancer therapeutics. Beyond oncology, this review also explores the expanded applications of cuproptosis, such as antibacterial, wound healing, and bone tissue engineering, highlighting its great potential to open innovative therapeutic strategies. Furthermore, the clinical potential of cuproptosis is assessed from basic, preclinical to clinical research. Finally, this review addresses current challenges, proposes potential solutions, and discusses the future prospects of this burgeoning field, highlighting cuproptosis nanomedicine as a highly promising alternative to current clinical therapeutics.
Collapse
Affiliation(s)
- Lijie Mao
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Ji Lu
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xinyu Wen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Zhiyi Song
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cai Sun
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuanru Zhao
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Fang Huang
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Si Chen
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Dongyang Jiang
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cheng Zhong
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Ke Li
- School of Materials Science and Engineering, Hainan University, Haikou 570228, China.
| | - Xiangyu Lu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Jianlin Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| |
Collapse
|
2
|
Cai X, Liu Y, Luo G, Yu Z, Jiang C, Xu C. Ultrasound-assisted immunotherapy for malignant tumour. Front Immunol 2025; 16:1547594. [PMID: 40433381 PMCID: PMC12106521 DOI: 10.3389/fimmu.2025.1547594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Malignant tumour represents a significant global public health concern. The advent of immunotherapy has brought about a revolutionary shift in the landscape of tumour treatment, offering a ray of hope to patients across the globe. Immunotherapy strategies have demonstrated considerable promise in clinical trials. However, the immunosuppressive environment within the tumour microenvironment has constituted a significant obstacle to the advancement of immunotherapies. It is therefore imperative to develop more efficacious and personalised approaches. The utilisation of non-invasive ultrasound-assisted immunotherapy represents a promising strategy. Ultrasound has the capacity to induce an immune response and stimulate other drugs to achieve a specific response, thereby reducing the toxic side effects of treatment and enhancing the outcome of immunotherapy. This paper presents a systematic introduction to the various mechanisms related to ultrasound and reviews the recent advancements of ultrasound-assisted tumour immunotherapy, including ultrasonic ablation, combined application with contrast agents, and sonodynamic therapy.
Collapse
Affiliation(s)
- Xiaowen Cai
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yujie Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Guosheng Luo
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Zhanwang Yu
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Cheng Jiang
- School of Applied Biology, Shenzhen City Polytechnic, Shenzhen, China
| | - Chuanshan Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Xiao X, Yang S, Jiang G, He S. Current views and trends of nanomaterials as vectors for gene delivery since the 21st century: a bibliometric analysis. Nanomedicine (Lond) 2025; 20:439-454. [PMID: 39878523 PMCID: PMC11875476 DOI: 10.1080/17435889.2025.2457781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Gene therapy is garnering increasing support due to its potential for a "once-delivered, lifelong benefit." The limitations of traditional gene delivery methods have spurred the advancement of bionanomaterials. Despite this progress, a thorough analysis of the evolution, current state, key contributors, focal studies, and future directions of nanomaterials in gene delivery remains absent. METHODS This study scrutinizes articles from the Web of Science, spanning 1 January 2 000, to 31 December 2023, employing various online tools for analysis and visualization. RESULTS The 21st century has witnessed consistent growth in scholarly work in this domain globally, with notable contributions from China and the US. At the same time, the Chinese Academy of Sciences (CAS), Harvard University, and Massachusetts Institute of Technology (MIT) have emerged as the most productive institutions, with CAS's academician Weihong Tan becoming the field's leading author. While drug delivery and nanoparticles (NPs) have been central themes for two decades, the research focus has shifted from modifying NPs and ultrafine particles to exploring polymer-hybrid NPs, mRNA vaccines, immune responses, green synthesis, and CRISPR/Cas tools. CONCLUSIONS This shift marks the transition from nanomaterials to bionanomaterials. The insights provided by this research offer a comprehensive overview of the field and valuable guidance for future investigations.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sheng Yang
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ge Jiang
- Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shisheng He
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Zhang A, Zhang X, Chen J, Shi X, Yu X, He Z, Sun J, Sun M, Liu Z. Approaches and applications in transdermal and transpulmonary gene drug delivery. Front Bioeng Biotechnol 2025; 12:1519557. [PMID: 39881959 PMCID: PMC11775749 DOI: 10.3389/fbioe.2024.1519557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Gene therapy has emerged as a pivotal component in the treatment of diverse genetic and acquired human diseases. However, effective gene delivery remains a formidable challenge to overcome. The presence of degrading enzymes, acidic pH conditions, and the gastrointestinal mucus layer pose significant barriers for genetic therapy, necessitating exploration of alternative therapeutic options. In recent years, transdermal and transpulmonary gene delivery modalities offer promising avenues with multiple advantages, such as non-invasion, avoided liver first-pass effect and improved patient compliance. Considering the rapid development of gene therapeutics via transdermal and transpulmonary administration, here we aim to summarize the nearest advances in transdermal and transpulmonary gene drug delivery. In this review, we firstly elaborate on current delivery carrier in gene therapy. We, further, describe approaches and applications for enhancing transdermal and transpulmonary gene delivery encompassing microneedles, chemical enhancers, physical methods for transdermal administration as well as nebulized formulations, dry powder formulations, and pressurized metered dose formulations for efficient transpulmonary delivery. Last but not least, the opportunities and outlooks of gene therapy through both administrated routes are highlighted.
Collapse
Affiliation(s)
- Anni Zhang
- Department of Ultrasound, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - Xuran Zhang
- Department of Orthopedics, Fuxin Center Hospital, Fuxin, Liaoning, China
| | - Jiahui Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xijuan Yu
- Department of Ultrasound, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning, China
| | - Zhijun Liu
- Department of Ultrasound, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Cao Y, Zhao X, Miao Y, Wang X, Deng D. How the Versatile Self-Assembly in Drug Delivery System to Afford Multimodal Cancer Therapy? Adv Healthc Mater 2025; 14:e2403715. [PMID: 39587000 DOI: 10.1002/adhm.202403715] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/04/2024] [Indexed: 11/27/2024]
Abstract
The rapid development of self-assembly technology during the past few decades has effectively addressed plenty of the issues associated with carrier-based drug delivery systems, such as low loading efficiency, complex fabrication processes, and inherent toxicity of carriers. The integration of nanoscale delivery systems with self-assembly techniques has enabled efficient and targeted self-administration of drugs, enhanced bioavailability, prolonged circulation time, and controllable drug release. Concurrently, the limitations of single-mode cancer treatment, including low bioavailability, poor therapeutic outcomes, and significant side effects, have highlighted the urgent need for multimodal combined antitumor therapies. Set against the backdrop of multimodal cancer therapy, this review summarizes the research progress and applications of a large number of self-assembled drug delivery platforms, including natural small molecule self-assembled, carrier-free self-assembled, amphiphilic polymer-based self-assembled, peptide-based self-assembled, and metal-based self-assembled nano drug delivery systems. This review particularly analyzes the latest advances in the application of self-assembled nano drug delivery platforms in combined antitumor therapies mediated by chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, and immunotherapy, providing innovative research insights for further optimization and expansion of self-assembled nano drug delivery systems in the clinical translation and development of antitumor combined therapy.
Collapse
Affiliation(s)
- Yuqi Cao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaomin Zhao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhang Miao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Dawei Deng
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
6
|
Zhao H, Du F, Xiang X, Tang Y, Feng Z, Wang Z, Rong X, Qiu L. Progress in application of nanomedicines for enhancing cancer sono-immunotherapy. ULTRASONICS SONOCHEMISTRY 2024; 111:107105. [PMID: 39427436 PMCID: PMC11533716 DOI: 10.1016/j.ultsonch.2024.107105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/22/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Cancer immunotherapy has significant potential as a cancer treatment since it boosts the immune system and prevents immune escape to get rid of or fight cancers. However, its clinical applicability is still limited because of the low response rate and immune-related side effects. Recently ultrasound has been shown to alter the tumor immune microenvironment, enhance the effectiveness of other antitumor therapies, and cause tumors to become more sensitive to immunotherapy, thus providing new insights into cancer treatment. Nanomedicines are also anticipated to have a positive impact on improving the immunological effects and enhancing ultrasound effect for cancer therapy. Therefore, designing effective nanomedicines enhanced ultrasound effect for augmenting sono-immunotherapy has been a pivot on anticancer therapy. In this review, the immunological impacts of various ultrasound therapeutic modalities, ultrasound parameters, and their underlying mechanisms are discussed. Moreover, we highlight the recent progress of nanomedicines synergistically enhancing sono-immunotherapy. Finally, we put forward opportunities and challenges on sono-immunotherapy.
Collapse
Affiliation(s)
- Hongxin Zhao
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fangxue Du
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xi Xiang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Tang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyan Feng
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyao Wang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiao Rong
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Li Qiu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Zhu M, Chen X, Zhang Y, Chen Y, Wu J, Duan X. Intestinal probiotic-based nanoparticles for cytotoxic siRNA delivery in immunotherapy against cancer. Int J Pharm 2024; 665:124689. [PMID: 39278289 DOI: 10.1016/j.ijpharm.2024.124689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/14/2024] [Accepted: 09/07/2024] [Indexed: 09/18/2024]
Abstract
Immunogene therapy has emerged as strategy against cancer by introducing immune-stimulating components into gene therapy. However, there is still a need for an ideal platform to achieve both immune stimulation and efficient gene delivery. Lactobacillus reuteri has potential immunomodulatory activity owing to its unique antigenicity, which is potentially relevant to cancer progression. Here, we designed a novel non-viral siRNA vector (DMPLAC) by encapsulating Lactobacillus reuteri lysate in DMP. DMPLAC can promote maturation and activation of immune cells, increase infiltration of APC and cytotoxic T cells in tumor microenvironment, and exhibit tumor suppressive effects. Loading of siRNA targeting Stat3, DMPLAC/siStat3 further inhibits tumor in multiple models. We designed a strategy that combines immune activation with Stat3 silencing, triggering an immune response and tumor killing. This dual-functional design provides a new choice in development of effective immunogene therapy.
Collapse
Affiliation(s)
- Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiaohua Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yueyang Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yang Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
8
|
Cheng M, Liu Y, You Q, Lei Z, Ji J, Zhang F, Dong WF, Li L. Metal-Doping Strategy for Carbon-Based Sonosensitizer in Sonodynamic Therapy of Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404230. [PMID: 38984451 PMCID: PMC11425966 DOI: 10.1002/advs.202404230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/25/2024] [Indexed: 07/11/2024]
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor and known for its challenging prognosis. Sonodynamic therapy (SDT) is an innovative therapeutic approach that shows promise in tumor elimination by activating sonosensitizers with low-intensity ultrasound. In this study, a novel sonosensitizer is synthesized using Cu-doped carbon dots (Cu-CDs) for the sonodynamic treatment of GBM. Doping with copper transforms the carbon dots into a p-n type semiconductor having a bandgap of 1.58 eV, a prolonged lifespan of 10.7 µs, and an improved electron- and hole-separation efficiency. The sonodynamic effect is efficiency enhanced. Western blot analysis reveals that the Cu-CDs induces a biological response leading to cell death, termed as cuproptosis. Specifically, Cu-CDs upregulate dihydrosulfanyl transacetylase expression, thereby establishing a synergistic therapeutic effect against tumor cell death when combined with SDT. Furthermore, Cu-CDs exhibit excellent permeability through the blood-brain barrier and potent anti-tumor activity. Importantly, the Cu-CDs effectively impede the growth of glioblastoma tumors and prolong the survival of mice bearing these tumors. This study provides support for the application of carbon-based nanomaterials as sonosensitizers in tumor therapy.
Collapse
Affiliation(s)
- Mingming Cheng
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| | - Yan Liu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| | - Qiannan You
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| | - Zhubing Lei
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| | - Jiajian Ji
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| | - Fan Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| | - Wen-Fei Dong
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| | - Li Li
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science (CAS), Suzhou, 215163, China
| |
Collapse
|
9
|
Zhou Y, Gao Y, Yao N, Lu G, Dong C, Wang K, Zhang J, Sun J, Li K, Li X. Multi-modal triggered-release sonodynamic/chemo/phototherapy synergistic nanocarriers for the treatment of colon cancer. Front Bioeng Biotechnol 2024; 12:1439883. [PMID: 39104624 PMCID: PMC11298370 DOI: 10.3389/fbioe.2024.1439883] [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: 05/28/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Most colon cancer patients are diagnosed at an advanced stage, with a grim prognosis. In clinical, various combination therapies have been employed to enhance the efficacy of colon cancer treatment. The essence of combined treatment is the judicious selection and combination of various treatment units. Phototherapy (PT), sonodynamic therapy (SDT), and chemotherapy are treatment modalities that rely on the active molecules to treat tumors, and have been demonstrated to synergistically enhance tumor treatment efficacy. However, the differences in the metabolism of active molecules and hypoxic microenvironment of tumors have limited the synergistic effects of the aforementioned methods. To address this significant issue, in this study, we utilized polydopamine (PDA) as the encapsulated material to form a rigid shell that contains the therapeutic molecules IR-780 and methotrexate (MTX) on the surface of perfluorohexane (PFH) microdroplets through self-assembling method to develop an SDT/chemotherapy/PT combined nanoparticles (SCP NPs). Transmission electron microscopy (TEM) revealed that the nanoparticles exhibited a hollow shell structure, with an average size of approximately 100 nm. SCP NPs have excellent stability and biocompatibility in both in vitro and in vivo. The absorption and emission spectrum of the loaded IR-780 did not exhibit any significant shift, and the photothermal temperature rose to 92°C. Their ultrasonic cavitation effect was good and their cell inhibitory effect of MTX was maintained. SCP NPs can achieve multi-modal triggered release through ultrasound, laser irradiation, and pH, ensuring a simultaneous accumulation of therapeutic molecules in the tumor area and effectively alleviating tumor hypoxia. Additionally, both the near-infrared fluorescence (NIF) signal and the ultrasonic cavitation signal of the nanoparticles can be utilized for tracking and monitoring treatment efficacy. Most notably, SCP NPs exhibited outstanding synergistic treatment effects at low intervention levels, resulting in a 67% cure rate of tumors. These results provide an experimental basis for developing the new clinical treatments for colon cancer.
Collapse
Affiliation(s)
- Yun Zhou
- College of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Yueyang Gao
- College of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Nannan Yao
- College of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Guozhi Lu
- College of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Chuyu Dong
- The Second College of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Kexin Wang
- The Second College of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Junfeng Zhang
- Xi’an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Jing Sun
- College of Medical Technology, Xi’an Medical University, Xi’an, China
| | - Ke Li
- Xi’an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Xueping Li
- College of Clinical Medicine, Xi’an Medical University, Xi’an, China
- Xi’an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| |
Collapse
|
10
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [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: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
11
|
Taghdiri M, Mussolino C. Viral and Non-Viral Systems to Deliver Gene Therapeutics to Clinical Targets. Int J Mol Sci 2024; 25:7333. [PMID: 39000440 PMCID: PMC11242246 DOI: 10.3390/ijms25137333] [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: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Clustered regularly interspersed short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology has revolutionized the field of gene therapy as it has enabled precise genome editing with unprecedented accuracy and efficiency, paving the way for clinical applications to treat otherwise incurable genetic disorders. Typically, precise genome editing requires the delivery of multiple components to the target cells that, depending on the editing platform used, may include messenger RNA (mRNA), protein complexes, and DNA fragments. For clinical purposes, these have to be efficiently delivered into transplantable cells, such as primary T lymphocytes or hematopoietic stem and progenitor cells that are typically sensitive to exogenous substances. This challenge has limited the broad applicability of precise gene therapy applications to those strategies for which efficient delivery methods are available. Electroporation-based methodologies have been generally applied for gene editing applications, but procedure-associated toxicity has represented a major burden. With the advent of novel and less disruptive methodologies to deliver genetic cargo to transplantable cells, it is now possible to safely and efficiently deliver multiple components for precise genome editing, thus expanding the applicability of these strategies. In this review, we describe the different delivery systems available for genome editing components, including viral and non-viral systems, highlighting their advantages, limitations, and recent clinical applications. Recent improvements to these delivery methods to achieve cell specificity represent a critical development that may enable in vivo targeting in the future and will certainly play a pivotal role in the gene therapy field.
Collapse
Affiliation(s)
- Maryam Taghdiri
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, 79106 Freiburg, Germany
- Ph.D. Program, Faculty of Biology, University of Freiburg, 79106 Freiburg, Germany
| | - Claudio Mussolino
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
12
|
Shen Q, Li Z, Wang Y, Meyer MD, De Guzman MT, Lim JC, Xiao H, Bouchard RR, Lu GJ. 50-nm Gas-Filled Protein Nanostructures to Enable the Access of Lymphatic Cells by Ultrasound Technologies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307123. [PMID: 38533973 PMCID: PMC11550859 DOI: 10.1002/adma.202307123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 03/14/2024] [Indexed: 03/28/2024]
Abstract
Ultrasound imaging and ultrasound-mediated gene and drug delivery are rapidly advancing diagnostic and therapeutic methods; however, their use is often limited by the need for microbubbles, which cannot transverse many biological barriers due to their large size. Here, the authors introduce 50-nm gas-filled protein nanostructures derived from genetically engineered gas vesicles(GVs) that are referred to as 50 nmGVs. These diamond-shaped nanostructures have hydrodynamic diameters smaller than commercially available 50-nm gold nanoparticles and are, to the authors' knowledge, the smallest stable, free-floating bubbles made to date. 50 nmGVs can be produced in bacteria, purified through centrifugation, and remain stable for months. Interstitially injected 50 nmGVs can extravasate into lymphatic tissues and gain access to critical immune cell populations, and electron microscopy images of lymph node tissues reveal their subcellular location in antigen-presenting cells adjacent to lymphocytes. The authors anticipate that 50 nmGVs can substantially broaden the range of cells accessible to current ultrasound technologies and may generate applications beyond biomedicine as ultrasmall stable gas-filled nanomaterials.
Collapse
Affiliation(s)
- Qionghua Shen
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Zongru Li
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Yixian Wang
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, 77005, USA
| | - Marc T De Guzman
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Janie C Lim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Han Xiao
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
- SynthX Center, Rice University, Houston, TX, 77005, USA
| | - Richard R Bouchard
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - George J Lu
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
- Department of BioSciences, Rice University, Houston, TX, 77005, USA
- Rice Synthetic Biology Institute, Rice University, Houston, TX, 77005, USA
| |
Collapse
|
13
|
Song Y, Dong QQ, Ni YK, Xu XL, Chen CX, Chen W. Nano-Proteolysis Targeting Chimeras (Nano-PROTACs) in Cancer Therapy. Int J Nanomedicine 2024; 19:5739-5761. [PMID: 38882545 PMCID: PMC11180470 DOI: 10.2147/ijn.s448684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules that have the capability to induce specific protein degradation. While playing a revolutionary role in effectively degrading the protein of interest (POI), PROTACs encounter certain limitations that impede their clinical translation. These limitations encompass off-target effects, inadequate cell membrane permeability, and the hook effect. The advent of nanotechnology presents a promising avenue to surmount the challenges associated with conventional PROTACs. The utilization of nano-proteolysis targeting chimeras (nano-PROTACs) holds the potential to enhance specific tissue accumulation, augment membrane permeability, and enable controlled release. Consequently, this approach has the capacity to significantly enhance the controllable degradation of target proteins. Additionally, they enable a synergistic effect by combining with other therapeutic strategies. This review comprehensively summarizes the structural basis, advantages, and limitations of PROTACs. Furthermore, it highlights the latest advancements in nanosystems engineered for delivering PROTACs, as well as the development of nano-sized PROTACs employing nanocarriers as linkers. Moreover, it delves into the underlying principles of nanotechnology tailored specifically for PROTACs, alongside the current prospects of clinical research. In conclusion, the integration of nanotechnology into PROTACs harbors vast potential in enhancing the anti-tumor treatment response and expediting clinical translation.
Collapse
Affiliation(s)
- Yue Song
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, People’s Republic of China
| | - Qing-Qing Dong
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Yi-Ke Ni
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Chao-Xiang Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Wei Chen
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
14
|
Wu Y, Li J, Shu L, Tian Z, Wu S, Wu Z. Ultrasound combined with microbubble mediated immunotherapy for tumor microenvironment. Front Pharmacol 2024; 15:1304502. [PMID: 38487163 PMCID: PMC10937735 DOI: 10.3389/fphar.2024.1304502] [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: 09/29/2023] [Accepted: 01/11/2024] [Indexed: 03/17/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in dynamically regulating the progress of cancer and influencing the therapeutic results. Targeting the tumor microenvironment is a promising cancer treatment method in recent years. The importance of tumor immune microenvironment regulation by ultrasound combined with microbubbles is now widely recognized. Ultrasound and microbubbles work together to induce antigen release of tumor cell through mechanical or thermal effects, promoting antigen presentation and T cells' recognition and killing of tumor cells, and improve tumor immunosuppression microenvironment, which will be a breakthrough in improving traditional treatment problems such as immune checkpoint blocking (ICB) and himeric antigen receptor (CAR)-T cell therapy. In order to improve the therapeutic effect and immune regulation of TME targeted tumor therapy, it is necessary to develop and optimize the application system of microbubble ultrasound for organs or diseases. Therefore, the combination of ultrasound and microbubbles in the field of TME will continue to focus on developing more effective strategies to regulate the immunosuppression mechanisms, so as to activate anti-tumor immunity and/or improve the efficacy of immune-targeted drugs, At present, the potential value of ultrasound combined with microbubbles in TME targeted therapy tumor microenvironment targeted therapy has great potential, which has been confirmed in the experimental research and application of breast cancer, colon cancer, pancreatic cancer and prostate cancer, which provides a new alternative idea for clinical tumor treatment. This article reviews the research progress of ultrasound combined with microbubbles in the treatment of tumors and their application in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Zuohui Wu
- Department of Ultrasound, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
15
|
Li J, Lu Z, Xu L, Wang J, Qian S, Hu Q, Ge Y. Poly(ethylenimine)-Cyclodextrin-Based Cationic Polymer Mediated HIF-1α Gene Delivery for Hindlimb Ischemia Treatment. ACS APPLIED BIO MATERIALS 2024; 7:1081-1094. [PMID: 38294873 DOI: 10.1021/acsabm.3c01020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Hindlimb ischemia is a common disease worldwide featured by the sudden decrease in limb perfusion, which usually causes a potential threat to limb viability and even amputation or death. Revascularization has been defined as the gold-standard therapy for hindlimb ischemia. Considering that vascular injury recovery requires cellular adaptation to the hypoxia, hypoxia-inducible factor 1 α (HIF-1α) is a potential gene for tissue restoration and angiogenesis. In this manuscript, effective gene delivery vector PEI-β-CD (PC) was reported for the first application in the hindlimb ischemia treatment to deliver HIF-1α plasmid in vitro and in vivo. Our in vitro finding demonstrated that PC/HIF-1α-pDNA could be successfully entered into the cells and mediated efficient gene transfection with good biocompatibility. More importantly, under hypoxic conditions, PC/HIF-1α-pDNA could up-regulate the HUEVC cell viability. In addition, the mRNA levels of VEGF, Ang-1, and PDGF were upregulated, and transcriptome results also demonstrated that the cell-related function of response to hypoxia was enhanced. The therapeutic effect of PC/HIF-1α-pDNA was further estimated in a murine acute hindlimb ischemia model, which demonstrated that intramuscular injection of PC/HIF-1α-pDNA resulted in significantly increased blood perfusion and alleviation in tissue damage, such as tissue fibrosis and inflammation. The results provide a rationale that HIF-1α-mediated gene therapy might be a practical strategy for the treatment of limb ischemia.
Collapse
Affiliation(s)
- Jingyu Li
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhuoting Lu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Liwang Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jing Wang
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 314408, China
| | - Shaojie Qian
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 314408, China
| | - Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yunfen Ge
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 314408, China
| |
Collapse
|
16
|
Vosough P, Vafadar A, Naderi S, Alashti SK, Karimi S, Irajie C, Savardashtaki A, Taghizadeh S. Escherichia coli cytosine deaminase: Structural and biotechnological aspects. Biotechnol Appl Biochem 2024; 71:5-16. [PMID: 37743549 DOI: 10.1002/bab.2516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/11/2023] [Indexed: 09/26/2023]
Abstract
Suicide gene therapy involves introducing viral or bacterial genes into tumor cells, which enables the conversion of a nontoxic prodrug into a toxic-lethal drug. The application of the bacterial cytosine deaminase (bCD)/5-fluorocytosine (5-FC) approach has been beneficial and progressive within the current field of cancer therapy because of the enhanced bystander effect. The basis of this method is the preferential deamination of 5-FC to 5-fluorouracil by cancer cells expressing cytosine deaminase (CD), which strongly inhibits DNA synthesis and RNA function, effectively targeting tumor cells. However, the poor binding affinity of toward 5-FC compared to the natural substrate cytosine and/or inappropriate thermostability limits the clinical applications of this gene therapy approach. Nowadays, many genetic engineering studies have been carried out to solve and improve the activity of this enzyme. In the current review, we intend to discuss the biotechnological aspects of Escherichia coli CD, including its structure, functions, molecular cloning, and protein engineering. We will also explore its relevance in cancer clinical trials. By examining these aspects, we hope to provide a thorough understanding of E. coli CD and its potential applications in cancer therapy.
Collapse
Affiliation(s)
- Parisa Vosough
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asma Vafadar
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samaneh Naderi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayan Khalili Alashti
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Karimi
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Cambyz Irajie
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Moradi Kashkooli F, Hornsby TK, Kolios MC, Tavakkoli JJ. Ultrasound-mediated nano-sized drug delivery systems for cancer treatment: Multi-scale and multi-physics computational modeling. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1913. [PMID: 37475577 DOI: 10.1002/wnan.1913] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 07/22/2023]
Abstract
Computational modeling enables researchers to study and understand various complex biological phenomena in anticancer drug delivery systems (DDSs), especially nano-sized DDSs (NSDDSs). The combination of NSDDSs and therapeutic ultrasound (TUS), that is, focused ultrasound and low-intensity pulsed ultrasound, has made significant progress in recent years, opening many opportunities for cancer treatment. Multiple parameters require tuning and optimization to develop effective DDSs, such as NSDDSs, in which mathematical modeling can prove advantageous. In silico computational modeling of ultrasound-responsive DDS typically involves a complex framework of acoustic interactions, heat transfer, drug release from nanoparticles, fluid flow, mass transport, and pharmacodynamic governing equations. Owing to the rapid development of computational tools, modeling the different phenomena in multi-scale complex problems involved in drug delivery to tumors has become possible. In the present study, we present an in-depth review of recent advances in the mathematical modeling of TUS-mediated DDSs for cancer treatment. A detailed discussion is also provided on applying these computational models to improve the clinical translation for applications in cancer treatment. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jahangir Jahan Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Shomope I, Percival KM, Abdel Jabbar NM, Husseini GA. Predicting Calcein Release from Ultrasound-Targeted Liposomes: A Comparative Analysis of Random Forest and Support Vector Machine. Technol Cancer Res Treat 2024; 23:15330338241296725. [PMID: 39539114 PMCID: PMC11561990 DOI: 10.1177/15330338241296725] [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: 07/03/2024] [Revised: 09/29/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE This study presents a comparative analysis of RF and SVM for predicting calcein release from ultrasound-triggered, targeted liposomes under varied low-frequency ultrasound (LFUS) power densities (6.2, 9, and 10 mW/cm2). METHODS Liposomes loaded with calcein and targeted with seven different moieties (cRGD, estrone, folate, Herceptin, hyaluronic acid, lactobionic acid, and transferrin) were synthesized using the thin-film hydration method. The liposomes were characterized using Dynamic Light Scattering and Bicinchoninic Acid assays. Extensive data collection and preprocessing were performed. RF and SVM models were trained and evaluated using mean absolute error (MAE), mean squared error (MSE), coefficient of determination (R²), and the a20 index as performance metrics. RESULTS RF consistently outperformed SVM, achieving R2 scores above 0.96 across all power densities, particularly excelling at higher power densities and indicating a strong correlation with the actual data. CONCLUSION RF outperforms SVM in drug release prediction, though both show strengths and apply based on specific prediction needs.
Collapse
Affiliation(s)
- Ibrahim Shomope
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Kelly M. Percival
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Nabil M. Abdel Jabbar
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah PO Box 26666, United Arab Emirates
| |
Collapse
|
19
|
Shi Y, Weng W, Chen M, Huang H, Chen X, Peng Y, Hu Y. Improving DNA vaccination performance through a new microbubble design and an optimized sonoporation protocol. ULTRASONICS SONOCHEMISTRY 2023; 101:106685. [PMID: 37976565 PMCID: PMC10692915 DOI: 10.1016/j.ultsonch.2023.106685] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/23/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
As a non-viral transfection method, ultrasound and microbubble-induced sonoporation can achieve spatially targeted gene delivery with synergistic immunostimulatory effects. Here, we report for the first time the application of sonoporation for improving DNA vaccination performance. This study developed a new microbubble design with nanoscale DNA/PEI complexes loaded onto cationic microbubbles to attain significant increases in DNA-loading capacity (0.25 pg per microbubble) and in vitro transfection efficiency. Using live-cell imaging, we revealed the membrane perforation and cellular delivery characteristics of sonoporation. Using luciferase reporter gene for in vivo transfection, we showed that sonoporation increased the transfection efficiency by 40.9-fold when compared with intramuscular injection. Moreover, we comprehensively optimized the sonoporation protocol and further increased the transfection efficiency by 43.6-fold. Immunofluorescent staining results showed that sonoporation effectively activated the MHC-II+ immune cells. Using a hepatitis B DNA vaccine, sonoporation induced significantly higher serum antibody levels when compared with intramuscular injection, and the antibodies sustained for 56 weeks. In addition, we recorded the longest reported expression period (400 days) of the sonoporation-delivered gene. Whole genome resequencing confirmed that the gene with stable expression existed in an extrachromosomal state without integration. Our results demonstrated the potential of sonoporation for efficient and safe DNA vaccination.
Collapse
Affiliation(s)
- Yuanchao Shi
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Weixiong Weng
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Mengting Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Haoqiang Huang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Xin Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Yin Peng
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Yaxin Hu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
20
|
Liu P, Foiret J, Situ Y, Zhang N, Kare AJ, Wu B, Raie MN, Ferrara KW, Qi LS. Sonogenetic control of multiplexed genome regulation and base editing. Nat Commun 2023; 14:6575. [PMID: 37852951 PMCID: PMC10584809 DOI: 10.1038/s41467-023-42249-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Manipulating gene expression in the host genome with high precision is crucial for controlling cellular function and behavior. Here, we present a precise, non-invasive, and tunable strategy for controlling the expression of multiple endogenous genes both in vitro and in vivo, utilizing ultrasound as the stimulus. By engineering a hyper-efficient dCas12a and effector under a heat shock promoter, we demonstrate a system that can be inducibly activated through thermal energy produced by ultrasound absorption. This system allows versatile thermal induction of gene activation or base editing across cell types, including primary T cells, and enables multiplexed gene activation using a single guide RNA array. In mouse models, localized temperature elevation guided by high-intensity focused ultrasound effectively triggers reporter gene expression in implanted cells. Our work underscores the potential of ultrasound as a clinically viable approach to enhance cell and gene-based therapies via precision genome and epigenome engineering.
Collapse
Affiliation(s)
- Pei Liu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Josquin Foiret
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Yinglin Situ
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Nisi Zhang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Aris J Kare
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Bo Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Marina N Raie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Katherine W Ferrara
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Osada T, Jiang X, Zhao Y, Chen M, Kreager BC, Wu H, Kim H, Ren J, Snyder J, Zhong P, Morse MA, Lyerly HK. The use of histotripsy as intratumoral immunotherapy beyond tissue ablation-the rationale for exploring the immune effects of histotripsy. Int J Hyperthermia 2023; 40:2263672. [PMID: 37806666 DOI: 10.1080/02656736.2023.2263672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
Mechanical high-intensity focused ultrasound (M-HIFU), which includes histotripsy, is a non-ionizing, non-thermal ablation technology that can be delivered by noninvasive methods. Because acoustic cavitation is the primary mechanism of tissue disruption, histotripsy is distinct from the conventional HIFU techniques resulting in hyperthermia and thermal injury. Phase I human trials have shown the initial safety and efficacy of histotripsy in treating patients with malignant liver tumors. In addition to tissue ablation, a promising benefit of M-HIFU has been stimulating a local and systemic antitumor immune response in preclinical models and potentially in the Phase I trial. Preclinical studies combining systemic immune therapies appear promising, but clinical studies of combinations have been complicated by systemic toxicities. Consequently, combining M-HIFU with systemic immunotherapy has been demonstrated in preclinical models and may be testing in future clinical studies. An additional alternative is to combine intratumoral M-HIFU and immunotherapy using microcatheter-placed devices to deliver both M-HIFU and immunotherapy intratumorally. The promise of M-HIFU as a component of anti-cancer therapy is promising, but as forms of HIFU are tested in preclinical and clinical studies, investigators should report not only the parameters of the energy delivered but also details of the preclinical models to enable analysis of the immune responses. Ultimately, as clinical trials continue, clinical responses and immune analysis of patients undergoing M-HIFU including forms of histotripsy will provide opportunities to optimize clinical responses and to optimize application and scheduling of M-HIFU in the context of the multi-modality care of the cancer patient.
Collapse
Affiliation(s)
- Takuya Osada
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Xiaoning Jiang
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | | | - Mengyue Chen
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | - Benjamin C Kreager
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | - Huaiyu Wu
- Department of Mechanical and Aerospace Engineering, College of Engineering, NC State University, Raleigh, NC, USA
| | - Howuk Kim
- Department of Mechanical Engineering, School of Engineering, Inha University, Incheon, Republic of South Korea
| | - Jun Ren
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Joshua Snyder
- Department of Surgery and Cell Biology, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Pei Zhong
- Thomas Lord Department of Mechanical Engineering and Material Science, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Michael A Morse
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, USA
| | - H Kim Lyerly
- Department of Surgery, Pathology, and Integrative Immunobiology, Duke University School of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
22
|
Wen Z, Liu C, Teng Z, Jin Q, Liao Z, Zhu X, Huo S. Ultrasound meets the cell membrane: for enhanced endocytosis and drug delivery. NANOSCALE 2023; 15:13532-13545. [PMID: 37548587 DOI: 10.1039/d3nr02562d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Endocytosis plays a crucial role in drug delivery for precision therapy. As a non-invasive and spatiotemporal-controllable stimulus, ultrasound (US) has been utilized for improving drug delivery efficiency due to its ability to enhance cell membrane permeability. When US meets the cell membrane, the well-known cavitation effect generated by US can cause various biophysical effects, facilitating the delivery of various cargoes, especially nanocarriers. The comprehension of recent progress in the biophysical mechanism governing the interaction between ultrasound and cell membranes holds significant implications for the broader scientific community, particularly in drug delivery and nanomedicine. This review will summarize the latest research results on the biological effects and mechanisms of US-enhanced cellular endocytosis. Moreover, the latest achievements in US-related biomedical applications will be discussed. Finally, challenges and opportunities of US-enhanced endocytosis for biomedical applications will be provided.
Collapse
Affiliation(s)
- Zihao Wen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Chen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zihao Teng
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Quanyi Jin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhihuan Liao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
23
|
Deng Q, Xie J, Kong S, Tang T, Zhou J. Long-Term Retention Microbubbles with Three-Layer Structure for Floating Intravesical Instillation Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205630. [PMID: 36634975 DOI: 10.1002/smll.202205630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/20/2022] [Indexed: 06/17/2023]
Abstract
Intravesical instillation is an effective treatment for bladder cancer. However, clinical anticancer agents always suffer rapid excretion by periodic urination, leading to low therapeutic efficacy. Prolonging the retention time of drugs in the bladder is the key challenge for intravesical instillation treatment. Herein, a facile and powerful surface cross-linking-freeze drying strategy is proposed to generate ultra-stable albumin bovine air microbubbles (BSA-MBs) that can float and adhere to the bladder wall to overcome the excretion of urination and exhibit a remarkable property of long-term retention in the bladder. More noteworthy, BSA-MBs are endowed with a specific three-layer structure, namely, the outer membrane, middle drug loading layer and inner air core, which makes them have a low density to easily float and possess a high drug loading capacity. Based on their unique superiorities, the therapeutic potential of doxorubicin (DOX)-loaded BSA-MBs (DOX-MBs) is exemplified by intravesical instillation for bladder cancer. After injection into the bladder, DOX-MBs can remain in the bladder for a long time and sustain the release of DOX in urine, exhibiting potent anticancer efficacy. Consequently, the prolonged retention of BSA-MBs in the bladder renders them as an effective floating drug delivery system for intravesical instillation therapy.
Collapse
Affiliation(s)
- Qiurong Deng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Junyi Xie
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shuying Kong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Tianmin Tang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jianhua Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
24
|
Hu X, Zhu H, He X, Chen J, Xiong L, Shen Y, Li J, Xu Y, Chen W, Liu X, Cao D, Xu X. The application of nanoparticles in immunotherapy for hepatocellular carcinoma. J Control Release 2023; 355:85-108. [PMID: 36708880 DOI: 10.1016/j.jconrel.2023.01.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/30/2023]
Abstract
Hepatocellular carcinoma (HCC) remains one of the leading causes of cancer-related deaths worldwide, however, current clinical diagnostic and treatment approaches remain relatively limited, creating an urgent need for the development of effective technologies. Immunotherapy has emerged as a powerful treatment strategy for advanced cancer. The number of clinically approved drugs for HCC immunotherapy has been increasing. However, it remains challenging to improve their transport and therapeutic efficiency, control their targeting and release, and mitigate their adverse effects. Nanotechnology has recently gained attention for improving the effectiveness of precision therapy for HCC. We summarize the key features of HCC associated with nanoparticle (NPs) targeting, release, and uptake, the roles and limitations of several major immunotherapies in HCC, the use of NPs in immunotherapy, the properties of NPs that influence their design and application, and current clinical trials of NPs in HCC, with the aim of informing the design of delivery platforms that have the potential to improve the safety and efficacy of HCC immunotherapy,and thus, ultimately improve the prognosis of HCC patients.
Collapse
Affiliation(s)
- Xinyao Hu
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaoqin He
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiayu Chen
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lin Xiong
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yang Shen
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiayi Li
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yangtao Xu
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenliang Chen
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xin Liu
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Dedong Cao
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Ximing Xu
- Cancer center, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
25
|
Hopkins C, Javius-Jones K, Wang Y, Hong H, Hu Q, Hong S. Combinations of chemo-, immuno-, and gene therapies using nanocarriers as a multifunctional drug platform. Expert Opin Drug Deliv 2022; 19:1337-1349. [PMID: 35949105 DOI: 10.1080/17425247.2022.2112569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Cancer immunotherapies have created a new generation of therapeutics to employ the immune system to attack cancer cells. However, these therapies are typically based on biologics that are nonspecific and often exhibit poor tumor penetration and dose-limiting toxicities. Nanocarriers allow the opportunity to overcome these barriers as they have the capabilities to direct immunomodulating drugs to tumor sites via passive and active targeting, decreasing potential adverse effects from nonspecific targeting. In addition, nanocarriers can be multifunctionalized to deliver multiple cancer therapeutics in a single drug platform, offering synergistic potential from co-delivery approaches. AREAS COVERED This review focuses on the delivery of cancer therapeutics using emerging nanocarriers to achieve synergistic results via co-delivery of immune-modulating components (i.e. chemotherapeutics, monoclonal antibodies, and genes). EXPERT OPINION Nanocarrier-mediated delivery of combinatorial immunotherapy creates the opportunity to fine-tune drug release while achieving superior tumor targeting and tumor cell death, compared to free drug counterparts. As these nanoplatforms are constantly improved upon, combinatorial immunotherapy will afford the greatest benefit to treat an array of tumor types while inhibiting cancer evasion pathways.
Collapse
Affiliation(s)
- Caroline Hopkins
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Heejoo Hong
- Department of Clinical Pharmacology & Therapeutics, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA.,Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Li L, Zhang X, Zhou J, Zhang L, Xue J, Tao W. Non-Invasive Thermal Therapy for Tissue Engineering and Regenerative Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107705. [PMID: 35475541 DOI: 10.1002/smll.202107705] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/11/2022] [Indexed: 06/14/2023]
Abstract
Owing to the development of nanotechnology and noninvasive treatment, thermal therapy in combination with external stimuli has been applied for tissue engineering and regenerative medicine (TERM), which has attracted more and more attention in recent years. In this review, the recent progress of applying a variety of non-invasive thermal therapeutic modalities for TERM, including photothermal therapy, magnetic thermotherapy, and ultrasound thermotherapy, as well as other thermal therapeutics are discussed. The parameters and conditions that need to be considered and regulated to realize a well-controlled thermal therapy for tissue regeneration are also discussed. Afterwards, the current concerns and challenges of putting thermal therapy into clinical applications are pointed out. At last, perspectives are provided for the future development directions, aiming to providing opportunities and a novel pathway for TERM.
Collapse
Affiliation(s)
- Longfei Li
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xiaodi Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, United States
| | - Jun Zhou
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, United States
| | - Liqun Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, United States
| |
Collapse
|
27
|
Yeingst TJ, Arrizabalaga JH, Hayes DJ. Ultrasound-Induced Drug Release from Stimuli-Responsive Hydrogels. Gels 2022; 8:554. [PMID: 36135267 PMCID: PMC9498906 DOI: 10.3390/gels8090554] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/16/2022] Open
Abstract
Stimuli-responsive hydrogel drug delivery systems are designed to release a payload when prompted by an external stimulus. These platforms have become prominent in the field of drug delivery due to their ability to provide spatial and temporal control for drug release. Among the different external triggers that have been used, ultrasound possesses several advantages: it is non-invasive, has deep tissue penetration, and can safely transmit acoustic energy to a localized area. This review summarizes the current state of understanding about ultrasound-responsive hydrogels used for drug delivery. The mechanisms of inducing payload release and activation using ultrasound are examined, along with the latest innovative formulations and hydrogel design strategies. We also report on the most recent applications leveraging ultrasound activation for both cancer treatment and tissue engineering. Finally, the future perspectives offered by ultrasound-sensitive hydrogels are discussed.
Collapse
Affiliation(s)
- Tyus J. Yeingst
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
| | - Julien H. Arrizabalaga
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
| | - Daniel J. Hayes
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
- Materials Research Institute, Millennium Science Complex, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
- The Huck Institute of the Life Sciences, Millennium Science Complex, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
| |
Collapse
|
28
|
Bendjador H, Foiret J, Wodnicki R, Stephens DN, Krut Z, Park EY, Gazit Z, Gazit D, Pelled G, Ferrara KW. A theranostic 3D ultrasound imaging system for high resolution image-guided therapy. Theranostics 2022; 12:4949-4964. [PMID: 35836805 PMCID: PMC9274734 DOI: 10.7150/thno.71221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/14/2022] [Indexed: 01/12/2023] Open
Abstract
Microbubble contrast agents are a diagnostic tool with broad clinical impact and an increasing number of indications. Many therapeutic applications have also been identified. Yet, technologies for ultrasound guidance of microbubble-mediated therapy are limited. In particular, arrays that are capable of implementing and imaging microbubble-based therapy in three dimensions in real-time are lacking. We propose a system to perform and monitor microbubble-based therapy, capable of volumetric imaging over a large field-of-view. To propel the promise of the theranostic treatment strategies forward, we have designed and tested a unique array and system for 3D ultrasound guidance of microbubble-based therapeutic protocols based on the frequency, temporal and spatial requirements. Methods: Four 256-channel plane wave scanners (Verasonics, Inc, WA, USA) were combined to control a 1024-element planar array with 1.3 and 2.5 MHz therapeutic and imaging transmissions, respectively. A transducer aperture of ~40×15 mm was selected and Field II was applied to evaluate the point spread function. In vitro experiments were performed on commercial and custom phantoms to assess the spatial resolution, image contrast and microbubble-enhanced imaging capabilities. Results: We found that a 2D array configuration with 64 elements separated by λ-pitch in azimuth and 16 elements separated by 1.5λ-pitch in elevation ensured the required flexibility. This design, of 41.6 mm × 16 mm, thus provided both an extended field-of-view, up to 11 cm x 6 cm at 10 cm depth and steering of ±18° in azimuth and ±12° in elevation. At a depth of 16 cm, we achieved a volume imaging rate of 60 Hz, with a contrast ratio and resolution, respectively, of 19 dB, 0.8 mm at 3 cm and 20 dB and 2.1 mm at 12.5 cm. Conclusion: A single 2D array for both imaging and therapeutics, integrated with a 1024 channel scanner can guide microbubble-based therapy in volumetric regions of interest.
Collapse
Affiliation(s)
| | | | | | | | - Zoe Krut
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Zulma Gazit
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dan Gazit
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gadi Pelled
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | |
Collapse
|
29
|
Kim C, Lim M, Woodworth GF, Arvanitis CD. The roles of thermal and mechanical stress in focused ultrasound-mediated immunomodulation and immunotherapy for central nervous system tumors. J Neurooncol 2022; 157:221-236. [PMID: 35235137 PMCID: PMC9119565 DOI: 10.1007/s11060-022-03973-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/16/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND Focused ultrasound (FUS) is an emerging technology, offering the capability of tuning and prescribing thermal and mechanical treatments within the brain. While early works in utilizing this technology have mainly focused on maximizing the delivery of therapeutics across the blood-brain barrier (BBB), the potential therapeutic impact of FUS-induced controlled thermal and mechanical stress to modulate anti-tumor immunity is becoming increasingly recognized. OBJECTIVE To better understand the roles of FUS-mediated thermal and mechanical stress in promoting anti-tumor immunity in central nervous system tumors, we performed a comprehensive literature review on focused ultrasound-mediated immunomodulation and immunotherapy in brain tumors. METHODS First, we summarize the current clinical experience with immunotherapy. Then, we discuss the unique and distinct immunomodulatory effects of the FUS-mediated thermal and mechanical stress in the brain tumor-immune microenvironment. Finally, we highlight recent findings that indicate that its combination with immune adjuvants can promote robust responses in brain tumors. RESULTS Along with the rapid advancement of FUS technologies into recent clinical trials, this technology through mild-hyperthermia, thermal ablation, mechanical perturbation mediated by microbubbles, and histotripsy each inducing distinct vascular and immunological effects, is offering the unique opportunity to improve immunotherapeutic trafficking and convert immunologically "cold" tumors into immunologically "hot" ones that are prone to generate prolonged anti-tumor immune responses. CONCLUSIONS While FUS technology is clearly accelerating concepts for new immunotherapeutic combinations, additional parallel efforts to detail rational therapeutic strategies supported by rigorous preclinical studies are still in need to leverage potential synergies of this technology with immune adjuvants. This work will accelerate the discovery and clinical implementation of new effective FUS immunotherapeutic combinations for brain tumor patients.
Collapse
Affiliation(s)
- Chulyong Kim
- School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael Lim
- Department of Neurosurgery, School of Medicine (Oncology), of Neurology, of Otolaryngology, and of Radiation Oncology, Stanford University, Paulo Alto, CA, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA
| | - Costas D Arvanitis
- School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|