1
|
Qi Z, Gu J, Qu L, Shi X, He Z, Sun J, Tan L, Sun M. Advancements of engineered live oncolytic biotherapeutics (microbe/virus/cells): Preclinical research and clinical progress. J Control Release 2024; 375:209-235. [PMID: 39244159 DOI: 10.1016/j.jconrel.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The proven efficacy of immunotherapy in fighting tumors has been firmly established, heralding a new era in harnessing both the innate and adaptive immune systems for cancer treatment. Despite its promise, challenges such as inefficient delivery, insufficient tumor penetration, and considerable potential toxicity of immunomodulatory agents have impeded the advancement of immunotherapies. Recent endeavors in the realm of tumor prophylaxis and management have highlighted the use of living biological entities, including bacteria, oncolytic viruses, and immune cells, as a vanguard for an innovative class of live biotherapeutic products (LBPs). These LBPs are gaining recognition for their inherent ability to target tumors. However, these LBPs must contend with significant barriers, including robust immune clearance mechanisms, cytotoxicity and other in vivo adverse effects. Priority must be placed on enhancing their safety and therapeutic indices. This review consolidates the latest preclinical research and clinical progress pertaining to the exploitation of engineered biologics, spanning bacteria, oncolytic viruses, immune cells, and summarizes their integration with combination therapies aimed at circumventing current clinical impasses. Additionally, the prospective utilities and inherent challenges of the biotherapeutics are deliberated, with the objective of accelerating their clinical application in the foreseeable future.
Collapse
Affiliation(s)
- Zhengzhuo Qi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Junmou Gu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihang Qu
- The 4th People's Hospital of Shenyang, China Medical University, Shenyang, Liaoning, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 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, 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, China
| | - Lingchen Tan
- School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, 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, China.
| |
Collapse
|
2
|
Liu K, Kong L, Cui H, Zhang L, Xin Q, Zhuang Y, Guo C, Yao Y, Tao J, Gu X, Jiang C, Wu J. Thymosin α1 reverses oncolytic adenovirus-induced M2 polarization of macrophages to improve antitumor immunity and therapeutic efficacy. Cell Rep Med 2024; 5:101751. [PMID: 39357524 PMCID: PMC11513825 DOI: 10.1016/j.xcrm.2024.101751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 06/29/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
Although oncolytic adenoviruses are widely studied for their direct oncolytic activity and immunomodulatory role in cancer immunotherapy, the immunosuppressive feedback loop induced by oncolytic adenoviruses remains to be studied. Here, we demonstrate that type V adenovirus (ADV) induces the polarization of tumor-associated macrophages (TAMs) to the M2 phenotype and increases the infiltration of regulatory T cells (Tregs) in the tumor microenvironment (TME). By selectively compensating for these deficiencies, thymosin alpha 1 (Tα1) reprograms "M2-like" TAMs toward an antitumoral phenotype, thereby reprogramming the TME into a state more beneficial for antitumor immunity. Moreover, ADVTα1 is constructed by harnessing the merits of all the components for the aforementioned combinatorial therapy. Both exogenously supplied and adenovirus-produced Tα1 orchestrate TAM reprogramming and enhance the antitumor efficacy of ADV via CD8+ T cells, showing promising prospects for clinical translation. Our findings provide inspiration for improving oncolytic adenovirus combination therapy and designing oncolytic engineered adenoviruses.
Collapse
Affiliation(s)
- Kua Liu
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Lingkai Kong
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Huawei Cui
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Louqian Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Qilei Xin
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan, China
| | - Yan Zhuang
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Ciliang Guo
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | - Yongzhong Yao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Jinqiu Tao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, China
| | - Xiaosong Gu
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan, China.
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China; Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan, China.
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Medical School & School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China; Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan, China.
| |
Collapse
|
3
|
Wang G, Mu M, Zhang Z, Chen Y, Yang N, Zhong K, Li Y, Lu F, Guo G, Tong A. Systemic delivery of tannic acid-ferric-masked oncolytic adenovirus reprograms tumor microenvironment for improved therapeutic efficacy in glioblastoma. Cancer Gene Ther 2024:10.1038/s41417-024-00839-8. [PMID: 39385009 DOI: 10.1038/s41417-024-00839-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024]
Abstract
Glioblastoma (GBM) represents the most aggressive primary brain tumor, and urgently requires effective treatments. Oncolytic adenovirus (OA) shows promise as a potential candidate for clinical antitumor therapy, including in the treatment of GBM. Nevertheless, the systemic delivery of OA continues to face challenges, leading to significantly compromised antitumor efficacy. In this study, we developed an innovative approach by encapsulating CXCL11-armed OA with tannic acid and Fe3+ (TA-Fe3+) to realize the systemic delivery of OA. The nanocarrier's ability to protect the OA from elimination by host immune response was evaluated in vitro and in vivo. We evaluated the antitumor effect and safety profile of OA@TA-Fe3+ in a GBM-bearing mice model. OA@TA-Fe3+ effectively safeguarded the virus from host immune clearance and extended its circulation in vivo. After targeting tumor sites, TA-Fe3+ could dissolve and release Fe3+ and OA. Fe3+-induced O2 production from H2O2 relieved the hypoxic state, and promoted OA replication, leading to a remarkable alteration of tumor immune microenvironment and enhancement in antitumor efficacy. Moreover, the systemic delivery of OA@TA-Fe3+ was safe without inflammation or organ damage. Our findings demonstrated the promising potential of systemically delivering the engineered OA for effective oncolytic virotherapy against GBM.
Collapse
Affiliation(s)
- Guoqing Wang
- Department of Ophthalmology, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Min Mu
- Laboratory of Liquid Biopsy and Single Cell Research, Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yongdong Chen
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Nian Yang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Kunhong Zhong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yanfang Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Fang Lu
- Department of Ophthalmology, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China.
| | - Gang Guo
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
4
|
Wu L, Yang L, Qian X, Hu W, Wang S, Yan J. Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment. J Funct Biomater 2024; 15:229. [PMID: 39194667 DOI: 10.3390/jfb15080229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
With the rapid development of tumor immunotherapy, nanoparticle vaccines have attracted much attention as potential therapeutic strategies. A systematic review and analysis must be carried out to investigate the effect of mannose modification on the immune response to nanoparticles in regulating the tumor microenvironment, as well as to explore its potential clinical application in tumor therapy. Despite the potential advantages of nanoparticle vaccines in immunotherapy, achieving an effective immune response in the tumor microenvironment remains a challenge. Tumor immune escape and the overexpression of immunosuppressive factors limit its clinical application. Therefore, our review explored how to intervene in the immunosuppressive mechanism in the tumor microenvironment through the use of mannan-decorated lipid calcium phosphate nanoparticle vaccines to improve the efficacy of immunotherapy in patients with tumors and to provide new ideas and strategies for the field of tumor therapy.
Collapse
Affiliation(s)
- Liusheng Wu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 19077, Singapore
| | - Lei Yang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xinye Qian
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wang Hu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shuang Wang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
5
|
Cao Z, Liu J. Surface nanocoating of bacteria as a versatile platform to develop living therapeutics. Nat Protoc 2024:10.1038/s41596-024-01019-6. [PMID: 39044001 DOI: 10.1038/s41596-024-01019-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/01/2024] [Indexed: 07/25/2024]
Abstract
Bacteria have been extensively utilized as living therapeutics for disease treatment due to their unique characteristics, such as genetic manipulability, rapid proliferation and specificity to target disease sites. Various in vivo insults can, however, decrease the vitality of dosed bacteria, leading to low overall bioavailability. Additionally, the innate antigens on the bacterial surface and the released toxins and metabolites may cause undesired safety issues. These limitations inevitably result in inadequate treatment outcomes, thereby hindering the clinical transformation of living bacterial therapeutics. Recently, we have developed a versatile platform to prepare advanced living bacterial therapeutics by nanocoating bacteria individually via either chemical decoration or physical encapsulation, which can improve bioavailability and reduce side effects for enhanced microbial therapy. Here we use interfacial self-assembly to prepare lipid membrane-coated bacteria (LCB), exhibiting increased resistance against a variety of harsh environmental conditions owing to the nanocoating's protective capability. Meanwhile, we apply mechanical extrusion to generate cell membrane-coated bacteria (CMCB), displaying improved biocompatibility owing to the nanocoating's shielding effect. We describe their detailed preparation procedures and demonstrate the expected functions of the coated bacteria. We also show that following oral delivery and intravenous injection in mouse models, LCB and CMCB present appealing potential for treating colitis and tumors, respectively. Compared with bioengineering that lacks versatile molecular tools for heterogeneous expression, the surface nanocoating technique is convenient to introduce functional components without restriction on bacterial strain types. Excluding bacterial culture, the fabrication of LCB takes ~2 h, while the preparation of CMCB takes ~5 h.
Collapse
Affiliation(s)
- Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Ding J, Su R, Yang R, Xu J, Liu X, Yao T, Li S, Wang C, Zhang H, Yue Q, Zhan C, Li C, Gao X. Enhancing the Antitumor Efficacy of Oncolytic Adenovirus Through Sonodynamic Therapy-Augmented Virus Replication. ACS NANO 2024; 18:18282-18298. [PMID: 38953884 DOI: 10.1021/acsnano.4c01115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The therapeutic efficacy of oncolytic adenoviruses (OAs) relies on efficient viral transduction and replication. However, the limited expression of coxsackie-adenovirus receptors in many tumors, along with the intracellular antiviral signaling, poses significant obstacles to OA infection and oncolysis. Here, we present sonosensitizer-armed OAs (saOAs) that potentiate the antitumor efficacy of oncolytic virotherapy through sonodynamic therapy-augmented virus replication. The saOAs could not only efficiently infect tumor cells via transferrin receptor-mediated endocytosis but also exhibit enhanced viral replication and tumor oncolysis under ultrasound irradiation. We revealed that the sonosensitizer loaded on the viruses induced the generation of ROS within tumor cells, which triggered JNK-mediated autophagy, ultimately leading to the enhanced viral replication. In mouse models of malignant melanoma, the combination of saOAs and sonodynamic therapy elicited a robust antitumor immune response, resulting in significant inhibition of melanoma growth and improved host survival. This work highlights the potential of sonodynamic therapy in enhancing the effectiveness of OAs and provides a promising platform for fully exploiting the antitumor efficacy of oncolytic virotherapy.
Collapse
Affiliation(s)
- Junqiang Ding
- School of Pharmacy, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Runping Su
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Rong Yang
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Jinliang Xu
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Xiaoxiao Liu
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Tingting Yao
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Sha Li
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Cong Wang
- School of Pharmacy, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Hanchang Zhang
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Qi Yue
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Urumqi Middle Road, Shanghai 200040, China
| | - Changyou Zhan
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Cong Li
- School of Pharmacy, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xihui Gao
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| |
Collapse
|
7
|
Jia J, Wang X, Lin X, Zhao Y. Engineered Microorganisms for Advancing Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313389. [PMID: 38485221 DOI: 10.1002/adma.202313389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Engineered microorganisms have attracted significant interest as a unique therapeutic platform in tumor treatment. Compared with conventional cancer treatment strategies, engineering microorganism-based systems provide various distinct advantages, such as the intrinsic capability in targeting tumors, their inherent immunogenicity, in situ production of antitumor agents, and multiple synergistic functions to fight against tumors. Herein, the design, preparation, and application of the engineered microorganisms for advanced tumor therapy are thoroughly reviewed. This review presents a comprehensive survey of innovative tumor therapeutic strategies based on a series of representative engineered microorganisms, including bacteria, viruses, microalgae, and fungi. Specifically, it offers extensive analyses of the design principles, engineering strategies, and tumor therapeutic mechanisms, as well as the advantages and limitations of different engineered microorganism-based systems. Finally, the current challenges and future research prospects in this field, which can inspire new ideas for the design of creative tumor therapy paradigms utilizing engineered microorganisms and facilitate their clinical applications, are discussed.
Collapse
Affiliation(s)
- Jinxuan Jia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaocheng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| |
Collapse
|
8
|
Zeng M, Zhang W, Li Y, Yu L. Harnessing adenovirus in cancer immunotherapy: evoking cellular immunity and targeting delivery in cell-specific manner. Biomark Res 2024; 12:36. [PMID: 38528632 DOI: 10.1186/s40364-024-00581-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/09/2024] [Indexed: 03/27/2024] Open
Abstract
Recombinant adenovirus (rAd) regimens, including replication-competent oncolytic adenovirus (OAV) and replication-deficient adenovirus, have been identified as potential cancer therapeutics. OAV presents advantages such as selective replication, oncolytic efficacy, and tumor microenvironment (TME) remodeling. In this perspective, the principles and advancements in developing OAV toolkits are reviewed. The burgeoning rAd may dictate efficacy of conventional cancer therapies as well as cancer immunotherapies, including cancer vaccines, synergy with adoptive cell therapy (ACT), and TME reshaping. Concurrently, we explored the potential of rAd hitchhiking to adoptive immune cells or stem cells, highlighting how this approach facilitates synergistic interactions between rAd and cellular therapeutics at tumor sites. Results from preclinical and clinical trials in which immune and stem cells were infected with rAd have been used to address significant oncological challenges, such as postsurgical residual tumor tissue and metastatic tissue. Briefly, rAd can eradicate tumors through various mechanisms, resulting from tumor immunogenicity, reprogramming of the TME, enhancement of cellular immunity, and effective tumor targeting. In this context, we argue that rAd holds immense potential for enhancing cellular immunity and synergistically improving antitumor effects in combination with novel cancer immunotherapies.
Collapse
Affiliation(s)
- Miao Zeng
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Wei Zhang
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yisheng Li
- Shenzhen Haoshi Biotechnology Co., Ltd. No, 155 Hongtian Road, Xinqiao Street, Bao'an District, Shenzhen, Guangdong, 518125, China.
| | - Li Yu
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| |
Collapse
|
9
|
Chen Y, Chen X, Bao W, Liu G, Wei W, Ping Y. An oncolytic virus-T cell chimera for cancer immunotherapy. Nat Biotechnol 2024:10.1038/s41587-023-02118-7. [PMID: 38336902 DOI: 10.1038/s41587-023-02118-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 12/21/2023] [Indexed: 02/12/2024]
Abstract
The efficacy of oncolytic adenoviruses (OAs) for cancer therapy has been limited by insufficient delivery to tumors after systemic injection and the propensity of OAs to induce the expression of immune checkpoints. To address these limitations, we use T cells to deliver OAs into tumors and engineer the OA to express a Cas9 system targeting the PDL1 gene encoding the immune checkpoint protein PD-L1. By cloaking OAs with cell membranes presenting T cell-specific antigens, we physically conjugated OAs onto T cell surfaces by antigen-receptor interaction. We tested the oncolytic virus-T cell chimera (ONCOTECH) via intravenous delivery in mouse cancer models, including models of melanoma, pancreatic adenocarcinoma, lung cancer and glioblastoma. In the melanoma model, the in vivo delivery of ONCOTECH resulted in a strong accumulation of OAs in tumor cells, where PD-L1 expression was reduced by 50% and the single administration of ONCOTECH enabled 80% survival over 70 days. Collectively, ONCOTECH represents a promising translational technology to combine virotherapy and cell therapy.
Collapse
Affiliation(s)
- Yuxuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Xiaohong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Yi L, Ning Z, Xu L, Shen Y, Zhu X, Yu W, Xie J, Meng Z. The combination treatment of oncolytic adenovirus H101 with nivolumab for refractory advanced hepatocellular carcinoma: an open-label, single-arm, pilot study. ESMO Open 2024; 9:102239. [PMID: 38325225 PMCID: PMC10937204 DOI: 10.1016/j.esmoop.2024.102239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND H101, an innovative oncolytic adenovirus, has shown potential in modifying the tumor microenvironment from immunologically 'cold' to 'hot'. When combined with nivolumab, a programmed cell death protein 1 inhibitor, this synergy may offer substantial therapeutic benefits beyond the capabilities of each agent alone. PATIENTS AND METHODS In this pilot study, we assessed the efficacy and safety of combining H101 with nivolumab in advanced hepatocellular carcinoma (HCC) patients who failed prior systemic therapy. The participants received initial oncolytic virus (OV) pretreatment with intratumoral H101 injections (5.0 × 1011 vp/0.5 ml/vial, two vials per lesion) on days 1 and 3. Combination therapy started on day 8, with H101 administered every 2 or 4 weeks and nivolumab (240 mg) injections every 2 weeks. Treatment continued up to 12 months or until disease progression, intolerable toxicity, consent withdrawal, or study conclusion. The primary endpoint was the objective response rate (ORR). RESULTS Between March 2020 and March 2022, 18 of 21 screened patients were assessable, showing an ORR of 11.1% [two cases of partial response (PR) and five cases of stable disease], with extrahepatic injections often leading to favorable outcomes. The disease control rate stood at 38.9%, with a 6-month survival rate of 88.9%. Median progression-free survival was 2.69 months, and overall survival (OS) was 15.04 months. Common adverse events included low-grade fever (100%) and pain related to centesis (33.3%), and no grade 3/4 events were reported. Significantly, local H101 injection showed potential in reversing immune checkpoint inhibitor resistance, evidenced by over 2.5 years of extended OS in PR cases with low α-fetoprotein. Additionally, decreasing neutrophil-to-lymphocyte ratio during OV pretreatment may predict positive outcomes. CONCLUSIONS This study demonstrates the potential efficacy of combining H101 with nivolumab in treating refractory advanced HCC, with well-tolerated toxicities.
Collapse
Affiliation(s)
- L Yi
- Department of Integrative Oncology, Shanghai, China; Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Z Ning
- Department of Integrative Oncology, Shanghai, China; Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - L Xu
- Department of Integrative Oncology, Shanghai, China; Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Y Shen
- Department of Integrative Oncology, Shanghai, China; Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - X Zhu
- Department of Integrative Oncology, Shanghai, China; Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - W Yu
- Department of Integrative Oncology, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - J Xie
- Department of Integrative Oncology, Shanghai, China; Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Z Meng
- Department of Integrative Oncology, Shanghai, China; Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Jin L, Mao Z. Living virus-based nanohybrids for biomedical applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1923. [PMID: 37619605 DOI: 10.1002/wnan.1923] [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: 02/23/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023]
Abstract
Living viruses characterized by distinctive biological functions including specific targeting, gene invasion, immune modulation, and so forth have been receiving intensive attention from researchers worldwide owing to their promising potential for producing numerous theranostic modalities against diverse pathological conditions. Nevertheless, concerns during applications, such as rapid immune clearance, altering immune activation modes, insufficient gene transduction efficiency, and so forth, highlight the crucial issues of excessive therapeutic doses and the associated biosafety risks. To address these concerns, synthetic nanomaterials featuring unique physical/chemical properties are frequently exploited as efficient drug delivery vehicles or treatments in biomedical domains. By constant endeavor, researchers nowadays can create adaptable living virus-based nanohybrids (LVN) that not only overcome the limitations of virotherapy, but also combine the benefits of natural substances and nanotechnology to produce novel and promising therapeutic and diagnostic agents. In this review, we discuss the fundamental physiochemical properties of the viruses, and briefly outline the basic construction methodologies of LVN. We then emphasize their distinct diagnostic and therapeutic performances for various diseases. Furthermore, we survey the foreseeable challenges and future perspectives in this interdisciplinary area to offer insights. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
He X, Yao W, Zhu JD, Jin X, Liu XY, Zhang KJ, Zhao SL. Potent antitumor efficacy of human dental pulp stem cells armed with YSCH-01 oncolytic adenovirus. J Transl Med 2023; 21:688. [PMID: 37789452 PMCID: PMC10546667 DOI: 10.1186/s12967-023-04539-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Systemic administration of oncolytic adenovirus for cancer therapy is still a challenge. Mesenchymal stem cells as cell carriers have gained increasing attention in drug delivery due to their excellent tumor tropism, immunosuppressive modulatory effects, and paracrine effects. However, the potential of human dental pulp stem cells (hDPSCs) loaded with oncolytic adenovirus for cancer biotherapy has not been investigated yet. METHODS The stemness of hDPSCs was characterized by FACS analysis and Alizarin red staining, Oil Red O staining, and immunofluorescence assays. The biological fitness of hDPSCs loaded with oncolytic adenovirus YSCH-01 was confirmed by virus infection with different dosages and cell viability CCK-8 assays. Additionally, the expression of CAR receptor in hDPSCs was detected by qPCR assay. Tumor tropism of hDPSC loaded with YSCH-01 in vitro and in vivo was investigated by Transwell assays and living tumor-bearing mice imaging technology and immunohistochemistry, Panoramic scanning of frozen section slices assay analysis. Furthermore, the antitumor efficacy was observed through the different routes of YSCH-01/hPDSCs administration in SW780 and SCC152 xenograft models. The direct tumor cell-killing effect of YSCH-01/hDPSCs in the co-culture system was studied, and the supernatant of YSCH-01/hDPSCs inhibited cell growth was further analyzed by CCK-8 assays. RESULTS hDPSCs were found to be susceptible to infection by a novel oncolytic adenovirus named YSCH-01 and were capable of transporting this virus to tumor sites at 1000 VP/cell infectious dosage in vitro and in vivo. Moreover, it was discovered that intraperitoneal injection of hDPSCs loaded with oncolytic adenovirus YSCH-01 exhibited potential anti-tumor effects in both SW780 and SCC152 xenograft models. The crucial role played by the supernatant secretome derived from hDPSCs loaded with YSCH-01 significantly exerted a specific anti-tumor effect without toxicity for normal cells, in both an active oncolytic virus and an exogenous protein-independent manner. Furthermore, the use of hDPSCs as a cell carrier significantly reduced the required dosage of virus delivery in vivo compared to other methods. CONCLUSIONS These findings highlight the promising clinical potential of hDPSCs as a novel cell carrier in the field of oncolytic virus-based anti-cancer therapy.
Collapse
Affiliation(s)
- Xu He
- Department of Stomatology, Huashan Hospital, Fudan University, 12 Urumqi Road, Jing'an District, Shanghai, 200040, China
| | - Wei Yao
- Shanghai Fengxian Stomatological Hospital, 189 Wanghe Road, Fengxian District, Shanghai, 201499, China
| | - Ji-Ding Zhu
- Shanghai Fengxian Stomatological Hospital, 189 Wanghe Road, Fengxian District, Shanghai, 201499, China
| | - Xin Jin
- Department of Stomatology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, 160 Pujian Road, Pudong New Area, Shanghai, 200025, China
| | - Xin-Yuan Liu
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, 1588 Huhang Road, Fengxian District, Shanghai, 201499, China
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 320 Yueyang Road, Xuhui District, Shanghai, 200031, China
| | - Kang-Jian Zhang
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, 1588 Huhang Road, Fengxian District, Shanghai, 201499, China.
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, 928 Second Avenue, Xiasha Higher Education Zone, Hangzhou, 310018, China.
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 320 Yueyang Road, Xuhui District, Shanghai, 200031, China.
| | - Shou-Liang Zhao
- Department of Stomatology, Huashan Hospital, Fudan University, 12 Urumqi Road, Jing'an District, Shanghai, 200040, China.
| |
Collapse
|
13
|
Liao ZX, Hsu SH, Tang SC, Kempson I, Yang PC, Tseng SJ. Potential targeting of the tumor microenvironment to improve cancer virotherapy. Pharmacol Ther 2023; 250:108521. [PMID: 37657673 DOI: 10.1016/j.pharmthera.2023.108521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
In 2015, oncolytic virotherapy was approved for clinical use, and in 2017, recombinant adeno-associated virus (AAV) delivery was also approved. However, systemic administration remains challenging due to the limited number of viruses that successfully reach the target site. Although the US Food and Drug Administration (FDA) permits the use of higher doses of AAV to achieve greater rates of transduction, most AAV still accumulates in the liver, potentially leading to toxicity there and elsewhere. Targeting the tumor microenvironment is a promising strategy for cancer treatment due to the critical role of the tumor microenvironment in controlling tumor progression and influencing the response to therapies. Newly discovered evidence indicates that administration routes focusing on the tumor microenvironment can promote delivery specificity and transduction efficacy within the tumor. Here, we review approaches that involve modifying viral surface features, modulating the immune system, and targeting the physicochemical characteristics in tumor microenvironment to regulate therapeutic delivery. Targeting tumor acidosis presents advantages that can be leveraged to enhance virotherapy outcomes and to develop new therapeutic approaches that can be integrated with standard treatments.
Collapse
Affiliation(s)
- Zi-Xian Liao
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10051, Taiwan
| | - Shiue-Cheng Tang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - S Ja Tseng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan; National Taiwan University YongLin Institute of Health, National Taiwan University, Taipei 10051, Taiwan; Program in Precision Health and Intelligent Medicine, Graduate School of Advanced Technology, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
14
|
Chen C, Zhu Z. Recent Advances in the Nanoshells Approach for Encapsulation of Single Probiotics. Drug Des Devel Ther 2023; 17:2763-2774. [PMID: 37705759 PMCID: PMC10497064 DOI: 10.2147/dddt.s419897] [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: 05/04/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023] Open
Abstract
The intestine, often referred to as the "second brain" of the human body, houses a vast microbial community that plays a crucial role in maintaining the host's balance and directly impacting overall health. Probiotics, a type of beneficial microorganism, offer various health benefits when consumed. However, probiotics face challenges such as acidic conditions in the stomach, bile acids, enzymes, and other adverse factors before they can colonize the intestinal tissues. At present, pills, dry powder, encapsulation, chemically modified bacteria, and genetically engineered bacteria have emerged as the preferred method for the stable and targeted delivery of probiotics. In particular, the use of nanoshells on the surface of single probiotics has shown promise in regulating their growth and differentiation. These nanoshells can detach from the probiotics' surface upon reaching the intestine, facilitating direct contact between the probiotics and intestinal mucosa. In this perspective, we provide an overview of the current developments in the formation of nanoshells mediated by single probiotics. We also discuss the advantages and disadvantages of different nanocoating strategies and explore future trends in probiotic protection.
Collapse
Affiliation(s)
- Cheng Chen
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu Province, 212300, People’s Republic of China
| | - Ziyu Zhu
- The Affiliated Huai’an Hospital of Xuzhou Medical University and the Second People’s Hospital of Huai’an, Huai’an, 223002, People’s Republic of China
| |
Collapse
|
15
|
Sadri M, Najafi A, Rahimi A, Behranvand N, Hossein Kazemi M, Khorramdelazad H, Falak R. Hypoxia effects on oncolytic virotherapy in Cancer: Friend or Foe? Int Immunopharmacol 2023; 122:110470. [PMID: 37433246 DOI: 10.1016/j.intimp.2023.110470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/13/2023]
Abstract
Researchers have tried to find novel strategies for cancer treatment in the past decades. Among the utilized methods, administering oncolytic viruses (OVs) alone or combined with other anticancer therapeutic approaches has had promising outcomes, especially in solid tumors. Infecting the tumor cells by these viruses can lead to direct lysis or induction of immune responses. However, the immunosuppressive tumor microenvironment (TME) is considered a significant challenge for oncolytic virotherapy in treating cancer. Based on OV type, hypoxic conditions in the TME can accelerate or repress virus replication. Therefore, genetic manipulation of OVs or other molecular modifications to reduce hypoxia can induce antitumor responses. Moreover, using OVs with tumor lysis capability in the hypoxic TME may be an attractive strategy to overcome the limitations of the therapy. This review summarizes the latest information available in the field of cancer virotherapy and discusses the dual effect of hypoxia on different types of OVs to optimize available related therapeutic methods.
Collapse
Affiliation(s)
- Maryam Sadri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Behranvand
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Feng C, Tan P, Nie G, Zhu M. Biomimetic and bioinspired nano-platforms for cancer vaccine development. EXPLORATION (BEIJING, CHINA) 2023; 3:20210263. [PMID: 37933383 PMCID: PMC10624393 DOI: 10.1002/exp.20210263] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2023]
Abstract
The advent of immunotherapy has revolutionized the treating modalities of cancer. Cancer vaccine, aiming to harness the host immune system to induce a tumor-specific killing effect, holds great promises for its broad patient coverage, high safety, and combination potentials. Despite promising, the clinical translation of cancer vaccines faces obstacles including the lack of potency, limited options of tumor antigens and adjuvants, and immunosuppressive tumor microenvironment. Biomimetic and bioinspired nanotechnology provides new impetus for the designing concepts of cancer vaccines. Through mimicking the stealth coating, pathogen recognition pattern, tissue tropism of pathogen, and other irreplaceable properties from nature, biomimetic and bioinspired cancer vaccines could gain functions such as longstanding, targeting, self-adjuvanting, and on-demand cargo release. The specific behavior and endogenous molecules of each type of living entity (cell or microorganism) offer unique features to cancer vaccines to address specific needs for immunotherapy. In this review, the strategies inspired by eukaryotic cells, bacteria, and viruses will be overviewed for advancing cancer vaccine development. Our insights into the future cancer vaccine development will be shared at the end for expediting the clinical translation.
Collapse
Affiliation(s)
- Chenchao Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Peng Tan
- Klarman Cell ObservatoryBroad Institute of MIT and HarvardCambridgeUSA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
- GBA Research Innovation Institute for NanotechnologyGuangzhouChina
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
| |
Collapse
|
17
|
Li X, Sun X, Wang B, Li Y, Tong J. Oncolytic virus-based hepatocellular carcinoma treatment: Current status, intravenous delivery strategies, and emerging combination therapeutic solutions. Asian J Pharm Sci 2023; 18:100771. [PMID: 36896445 PMCID: PMC9989663 DOI: 10.1016/j.ajps.2022.100771] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/24/2022] [Accepted: 12/04/2022] [Indexed: 12/30/2022] Open
Abstract
Current treatments for advanced hepatocellular carcinoma (HCC) have limited success in improving patients' quality of life and prolonging life expectancy. The clinical need for more efficient and safe therapies has contributed to the exploration of emerging strategies. Recently, there has been increased interest in oncolytic viruses (OVs) as a therapeutic modality for HCC. OVs undergo selective replication in cancerous tissues and kill tumor cells. Strikingly, pexastimogene devacirepvec (Pexa-Vec) was granted an orphan drug status in HCC by the U.S. Food and Drug Administration (FDA) in 2013. Meanwhile, dozens of OVs are being tested in HCC-directed clinical and preclinical trials. In this review, the pathogenesis and current therapies of HCC are outlined. Next, we summarize multiple OVs as single therapeutic agents for the treatment of HCC, which have demonstrated certain efficacy and low toxicity. Emerging carrier cell-, bioengineered cell mimetic- or nonbiological vehicle-mediated OV intravenous delivery systems in HCC therapy are described. In addition, we highlight the combination treatments between oncolytic virotherapy and other modalities. Finally, the clinical challenges and prospects of OV-based biotherapy are discussed, with the aim of continuing to develop a fascinating approach in HCC patients.
Collapse
Affiliation(s)
- Xinguo Li
- The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaonan Sun
- The 4th People's Hospital of Shenyang, Shenyang 110031, China
| | - Bingyuan Wang
- The First Hospital of China Medical University, Shenyang 110001, China
| | - Yiling Li
- The First Hospital of China Medical University, Shenyang 110001, China
| | - Jing Tong
- The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
18
|
Nanoarchitectured assembly and surface of two-dimensional (2D) transition metal dichalcogenides (TMDCs) for cancer therapy. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
Samoranos KT, Krisiewicz AL, Karpinecz BC, Glover PA, Gale TV, Chehadeh C, Ashshan S, Koya R, Chung EY, Lim HL. pH Sensitive Erythrocyte-Derived Membrane for Acute Systemic Retention and Increased Infectivity of Coated Oncolytic Vaccinia Virus. Pharmaceutics 2022; 14:pharmaceutics14091810. [PMID: 36145558 PMCID: PMC9504069 DOI: 10.3390/pharmaceutics14091810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/01/2022] Open
Abstract
Oncolytic viruses have emerged as a promising modality in cancer treatment given their high synergy with highly efficient immune checkpoint inhibitors. However, their potency is limited by their rapid in vivo clearance. To overcome this, we coated oncolytic vaccinia viruses (oVV) with erythrocyte-derived membranes (EDMs), hypothesizing that they would not only remain in systemic circulation for longer as erythrocytes would when administered intravenously, but also respond to environmental pH cues due to their membrane surface sialic acid residues. For this, we developed a model based on DLVO theory to show that the acidic moieties on the surface of EDM confers it the ability to respond to pH-based stimuli. We corroborate our modeling results through in vitro cell culture models and show that EDM-coated oVV infects cancer cells faster under acidic conditions akin to the tumor microenvironment. When EDM-coated oVVs were intravenously injected into wild-type mice, they exhibited prolonged circulation at higher concentrations when compared to the unprocessed oVV. Furthermore, when EDM-coated oVV was directly injected into xenografted tumors, we observed that they were suppressed earlier than the tumors that received regular oVV, suggesting that the EDM coating does not hinder oVV infectivity. Overall, we found that EDM was able to serve as a multi-functional encapsulant that allowed the payload to remain in circulation at higher concentrations when administered intravenously while simultaneously exhibiting pH-responsive properties.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Richard Koya
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Eddie Y. Chung
- Coastar Therapeutics Inc., San Diego, CA 92121, USA
- Correspondence: (E.Y.C.); (H.L.L.)
| | - Han L. Lim
- Coastar Therapeutics Inc., San Diego, CA 92121, USA
- Correspondence: (E.Y.C.); (H.L.L.)
| |
Collapse
|
20
|
Li Y, Tang K, Zhang X, Pan W, Li N, Tang B. Tumor microenvironment responsive nanocarriers for gene therapy. Chem Commun (Camb) 2022; 58:8754-8765. [PMID: 35880654 DOI: 10.1039/d2cc02759c] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stimuli responsive nanocarriers are important non-viral gene carriers for gene therapy. We discuss the stimulus conditions and then highlight various stimuli responsive nanocarriers in the tumor microenvironment for cancer gene therapy. We hope that this review will inspire readers to develop more effective stimuli responsive nanocarriers for delivering genes.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Kun Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Xia Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
21
|
Huang L, Zhao H, Shan M, Chen H, Xu B, He Y, Zhao Y, Liu Z, Chen J, Xu Q. Oncolytic adenovirus H101 ameliorate the efficacy of anti-PD-1 monotherapy in colorectal cancer. Cancer Med 2022; 11:4575-4587. [PMID: 35762456 PMCID: PMC9741988 DOI: 10.1002/cam4.4845] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 04/14/2022] [Accepted: 05/04/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Immune checkpoint blockade therapy with anti-programmed cell death (PD)-1 antibodies provides therapeutic effect for many patients of various cancers but remains inadequate in colorectal cancer (CRC) patients. The present study aims to assess the efficacy of oncolytic adenovirus (OncoAd ) in enhancing the anti-PD-1 treatment of CRC. METHODS The estimating relative subsets of RNA transcripts algorithm was used for estimating the infiltrated immune cells in melanoma and CRC tissues. The efficacy of OncoAd with anti-PD-1 monotherapy was performed in a CT26 CRC mouse model in vivo. Flow cytometric analysis of peripheral blood and tumor tissues determined the difference anti-tumor immune efficacy of OncoAd with anti-PD-1 monotherapy. RESULTS The Cancer Genome Atlas database indicated that CD8+ T cells and regulatory T cells were significantly elevated in melanoma compared to CRC cohorts. Moreover, intratumor injection of oncolytic adenovirus enhanced T cell infiltration and decreased Treg percentages in the CT26 CRC colorectal cancer mouse model. Combinatorial OncoAd with anti-PD-1 antibody treatment markedly enhanced the anti-tumor efficacy of anti-PD-1 by significantly decreasing the tumor volume and reducing tumor growth in a CRC mouse model. To the end, OncoAd treatment increased the CD8/Treg ratio, indicating that OncoAd intratumor injection ameliorate the anti-tumor immune response of anti-PD-1 therapy. CONCLUSION The present study elucidates that OncoAd promotes intratumor T cell infiltration and improves anti-PD-1 immunotherapy, thereby providing a potent combinatorial therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Lili Huang
- Department of Oncology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina,Tongji University Cancer CenterShanghaiChina
| | - Huaxin Zhao
- Department of Oncology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina,Tongji University Cancer CenterShanghaiChina
| | | | - Hong Chen
- Department of Gastrointestinal SurgeryFujian Provincial HospitalFuzhouChina
| | - Bin Xu
- Department of General Surgery, Shanghai Tenth People's HospitalTongji University School of MedicineShangaiChina
| | - Yang He
- Department of Oncology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Yu Zhao
- Department of Oncology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina,Tongji University Cancer CenterShanghaiChina
| | - Zhuqing Liu
- Department of Oncology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina,Tongji University Cancer CenterShanghaiChina
| | - Jianhua Chen
- Department of Oncology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina,Tongji University Cancer CenterShanghaiChina
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina,Tongji University Cancer CenterShanghaiChina,Anhui Medical UniversityHeFeiChina
| |
Collapse
|
22
|
Dick TA, Sone ED, Uludağ H. Mineralized vectors for gene therapy. Acta Biomater 2022; 147:1-33. [PMID: 35643193 DOI: 10.1016/j.actbio.2022.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/01/2022]
Abstract
There is an intense interest in developing materials for safe and effective delivery of polynucleotides using non-viral vectors. Mineralization of organic templates has long been used to produce complex materials with outstanding biocompatibility. However, a lack of control over mineral growth has limited the applicability of mineralized materials to a few in vitro applications. With better control over mineral growth and surface functionalization, mineralized vectors have advanced significantly in recent years. Here, we review the recent progress in chemical synthesis, physicochemical properties, and applications of mineralized materials in gene therapy, focusing on structure-function relationships. We contrast the classical understanding of the mineralization mechanism with recent ideas of mineralization. A brief introduction to gene delivery is summarized, followed by a detailed survey of current mineralized vectors. The vectors derived from calcium phosphate are articulated and compared to other minerals with unique features. Advanced mineral vectors derived from templated mineralization and specialty coatings are critically analyzed. Mineral systems beyond the co-precipitation are explored as more complex multicomponent systems. Finally, we conclude with a perspective on the future of mineralized vectors by carefully demarcating the boundaries of our knowledge and highlighting ambiguous areas in mineralized vectors. STATEMENT OF SIGNIFICANCE: Therapy by gene-based medicines is increasingly utilized to cure diseases that are not alleviated by conventional drug therapy. Gene medicines, however, rely on macromolecular nucleic acids that are too large and too hydrophilic for cellular uptake. Without tailored materials, they are not functional for therapy. One emerging class of nucleic acid delivery system is mineral-based materials. The fact that they can undergo controlled dissolution with minimal footprint in biological systems are making them attractive for clinical use, where safety is utmost importance. In this submission, we will review the emerging synthesis technology and the range of new generation minerals for use in gene medicines.
Collapse
|
23
|
Edwardson TGW, Levasseur MD, Tetter S, Steinauer A, Hori M, Hilvert D. Protein Cages: From Fundamentals to Advanced Applications. Chem Rev 2022; 122:9145-9197. [PMID: 35394752 DOI: 10.1021/acs.chemrev.1c00877] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteins that self-assemble into polyhedral shell-like structures are useful molecular containers both in nature and in the laboratory. Here we review efforts to repurpose diverse protein cages, including viral capsids, ferritins, bacterial microcompartments, and designed capsules, as vaccines, drug delivery vehicles, targeted imaging agents, nanoreactors, templates for controlled materials synthesis, building blocks for higher-order architectures, and more. A deep understanding of the principles underlying the construction, function, and evolution of natural systems has been key to tailoring selective cargo encapsulation and interactions with both biological systems and synthetic materials through protein engineering and directed evolution. The ability to adapt and design increasingly sophisticated capsid structures and functions stands to benefit the fields of catalysis, materials science, and medicine.
Collapse
Affiliation(s)
| | | | - Stephan Tetter
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Angela Steinauer
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Mao Hori
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
24
|
Zhang X, Liu Q, Zhang T, Gao P, Wang H, Yao L, Huang J, Jiang S. Bone-targeted nanoplatform enables efficient modulation of bone tumor microenvironment for prostate cancer bone metastasis treatment. Drug Deliv 2022; 29:889-905. [PMID: 35285760 PMCID: PMC8928789 DOI: 10.1080/10717544.2022.2050845] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
As there is currently no effective therapy for patients with prostate cancer (PCa) bone metastasis, it was stringent to explore the relevant treatment strategies. Actually, the interaction between cancer cells and bone microenvironment plays important role in prostate cancer bone metastasis, especially the Sonic hedgehog protein (SHH) signaling in the bone microenvironment. The SHH promotes osteoblast maturation and osteoblast then secretes RANKL to induce osteoclastogenesis. Herein, this study develops bone-targeting calcium phosphate lipid hybrid nanoparticles (NPs) loaded with docetaxel (DTXL) and SHH siRNA for PCa bone metastasis treatment. For bone targeting purposes, the nanoplatform was modified with alendronate (ALN). (DTXL + siRNA)@NPs-ALN NPs effectively change the bone microenvironment by inhibiting the SHH paracrine and autocrine signaling, enhancing the anti-tumor effects of DTXL. Besides showing good in vitro cellular uptake, the NPs-ALN also inhibited tumor growth both in vitro and in vivo by inducing apoptosis, cell cycle arrest, and autophagy. This DDS comprised of (DTXL + siRNA)-loaded NPs provides an excellent strategy to treat PCa bone metastasis.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Postdoctoral of Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Pathology, Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Tingting Zhang
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Pei Gao
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Hui Wang
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Lu Yao
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Jingwen Huang
- The First Affiliated Hospital of Bengbu Medical College, Tumor Hospital Affiliated to Bengbu Medical College, Bengbu, China
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| |
Collapse
|
25
|
Iscaro A, Jones C, Forbes N, Mughal A, Howard FN, Janabi HA, Demiral S, Perrie Y, Essand M, Weglarz A, Cruz LJ, Lewis CE, Muthana M. Targeting circulating monocytes with CCL2-loaded liposomes armed with an oncolytic adenovirus. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102506. [PMID: 34875352 DOI: 10.1016/j.nano.2021.102506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 09/17/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022]
Abstract
Oncolytic viruses (OVs) selectively replicate in and destroy cancer cells resulting in anti-tumor immunity. However, clinical use remains a challenge because of virus clearance upon intravenous delivery. OV packaging using a nanomedicine approach could overcome this. Here we encapsulate an oncolytic adenovirus (Ad[I/PPT-E1A]) into CCL2-coated liposomes in order to exploit recruitment of CCR2-expressing circulating monocytes into tumors. We demonstrate successful encapsulation of Ad[I/PPT-E1A] into CCL2-coated liposomes that were preferentially taken up by CCR2-expressing monocytes. No complex-related toxicities were observed following incubation with prostate tumor cells and the encapsulation did not affect virus oncolytic activity in vitro. Furthermore, intravenous administration of our nanomedicine resulted in a significant reduction in tumor size and pulmonary metastasis in prostate cancer-bearing mice whereby a 1000-fold less virus was needed compared to Ad[I/PPT-E1A] alone. Taken together our data provide an opportunity to target OVs via circulation to inaccessible tumors using liposome-assisted drug delivery.
Collapse
Affiliation(s)
- Alessandra Iscaro
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Christian Jones
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Neil Forbes
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Amina Mughal
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | | | - Haider Al Janabi
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Secil Demiral
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Aleksandra Weglarz
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Luis J Cruz
- Department of Radiology, Division Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Claire E Lewis
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Munitta Muthana
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK.
| |
Collapse
|
26
|
Wang C, Wang X, Zhang W, Ma D, Li F, Jia R, Shi M, Wang Y, Ma G, Wei W. Shielding Ferritin with a Biomineralized Shell Enables Efficient Modulation of Tumor Microenvironment and Targeted Delivery of Diverse Therapeutic Agents. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107150. [PMID: 34897858 DOI: 10.1002/adma.202107150] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/27/2021] [Indexed: 05/23/2023]
Abstract
Ferritin (Fn) is considered a promising carrier for targeted delivery to tumors, but the successful application in vivo has not been fully achieved yet. Herein, strong evidence is provided that the Fn receptor is expressed in liver tissues, resulting in an intercept effect in regards to tumor delivery. Building on these observations, a biomineralization technology is rationally designed to shield Fn using a calcium phosphate (CaP) shell, which can improve the delivery performance by reducing Fn interception in the liver while re-exposing it in acidic tumors. Moreover, the selective dissolution of the CaP shell not only neutralizes the acidic microenvironment but also induces the intratumoral immunomodulation and calcification. Upon multiple cell line and patient-derived xenografts, it is demonstrated that the elaboration of the highly flexible Fn@CaP chassis by loading a chemotherapeutic drug into the Fn cavity confers potent antitumor effects, and additionally encapsulating a photosensitizer into the outer shell enables a combined chemo-photothermal therapy for complete suppression of advanced tumors. Altogether, these results support Fn@CaP as a new nanoplatform for efficient modulation of the tumor microenvironment and targeted delivery of diverse therapeutic agents.
Collapse
Affiliation(s)
- Changlong Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaojun Wang
- Department of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518039, P. R. China
| | - Wei Zhang
- Beijing National Laboratory for Molecular Engineering, College of Chemistry and Molecular Engineering and College of Engineering and BIC-ESAT, Peking University, Beijing, 100871, P. R. China
| | - Ding Ma
- Beijing National Laboratory for Molecular Engineering, College of Chemistry and Molecular Engineering and College of Engineering and BIC-ESAT, Peking University, Beijing, 100871, P. R. China
| | - Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Rongrong Jia
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, P. R. China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, P. R. China
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
27
|
Ding L, Gao Q, Xu Z, Cai L, Chen S, Zhang X, Cao P, Chen G. An Inter-Supplementary Biohybrid System Based on Natural Killer Cells for the Combinational Immunotherapy and Virotherapy of Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103470. [PMID: 34747156 PMCID: PMC8805568 DOI: 10.1002/advs.202103470] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/12/2021] [Indexed: 05/05/2023]
Abstract
Oncolytic adenoviruses (Ads) have gained great attention in cancer therapy because they cause direct cytolytic infection and indirectly induce antitumor immunity. However, their efficacy is compromised by host antiviral immune response, poor tumor delivery, and the immunosuppressive tumor microenvironment (TME). Here, a natural killer (NK) cell-mediated Ad delivery system (Ad@NK) is generated by harnessing the merits of the two components for combinational immunotherapy and virotherapy of cancer. In this biohybrid system, NK cells with a tumor-homing tropism act as bioreactors and shelters for the loading, protection, replication, amplification, and release of Ads, thereby leading to a highly efficient systemic tumor-targeted delivery. As feedback, Ad infection offers NK cells an enhanced antitumor immunity by activating type I interferon signaling in a STAT4-granzyme B-dependent manner. Moreover, it is found that the Ad@NK system can relieve immunosuppression in the TME by promoting the maturation of dendritic cells and the polarization of macrophages to M1 phenotype. Both in vitro and in vivo data indicate the excellent antitumor and antimetastatic functions of Ad@NKs by destroying tumor cells, inducing immunogenic cell death, and immunomodulating TME. This work provides a clinical basis for improved oncolytic virotherapy in combination with NK cell therapy based on the inter-supplementary biohybrid system.
Collapse
Affiliation(s)
- Li Ding
- College of Bioscience and BiotechnologyYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Qingqing Gao
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Institute of Comparative MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Zhuobin Xu
- Institute of Translational MedicineMedical CollegeYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Liangliang Cai
- Institute of Translational MedicineMedical CollegeYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Sujuan Chen
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Institute of Comparative MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Xinyue Zhang
- College of Bioscience and BiotechnologyYangzhou UniversityYangzhouJiangsu225009P. R. China
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingJiangsu210023P. R. China
| | - Gang Chen
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Institute of Comparative MedicineYangzhou UniversityYangzhouJiangsu225009P. R. China
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou UniversityYangzhouJiangsu225009P. R. China
| |
Collapse
|
28
|
Liu Y, Wang Y, Song S, Zhang H. Tumor Diagnosis and Therapy Mediated by Metal Phosphorus-Based Nanomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103936. [PMID: 34596931 DOI: 10.1002/adma.202103936] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/14/2021] [Indexed: 05/23/2023]
Abstract
Metal phosphorus-based nanomaterials (Metal-P NMs) including metal phosphate nanomaterials, metal phosphide nanomaterials, and metal-black phosphorus (Metal-BP) nanocomposite are widely used in the field of biomedicine owing to their excellent physical and chemical properties, biocompatibility, and biodegradability. In recent years, metal phosphate nanomaterials and Metal-BP nanocomposite acted as medicine delivery system have made breakthroughs in tumor diagnosis including magnetic resonance imaging, fluorescence imaging, photoacoustic imaging, nuclear imaging, and therapies including chemotherapy, gene therapy, photothermal therapy, photodynamic therapy, and radiation therapy. Metal phosphate nanomaterials have good biodegradability, especially calcium-based metal phosphate nanomaterials can be dissolved into nontoxic ions and participate in the metabolisms of normal organs. Compared with metal phosphate nanomaterials, metal phosphide nanomaterials have excellent optical, magnetic, and catalytic properties, which can be used as multifunctional diagnostic nanoplatforms and therapeutic agents for chemodynamic therapy, photothermal therapy, or immunotherapy. The latest developments in Metal-P NMs, covering the range of preparation methods and biological applications, such as serving as drug carriers, tumor diagnosis, and therapy, are focused. All in all, the current trends, key issues, future prospects and challenges of Metal-P NMs are concluded and discussed, which are important for the development of this research field and shining more lights on this direction.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yinghui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Shuyan Song
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
- Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
29
|
Munir MU, Salman S, Javed I, Bukhari SNA, Ahmad N, Shad NA, Aziz F. Nano-hydroxyapatite as a delivery system: overview and advancements. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2021; 49:717-727. [PMID: 34907839 DOI: 10.1080/21691401.2021.2016785] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nano-hydroxyapatite is being investigated as vital components of implants and dental and tissue engineering devices. It is found as a bone replacement due to its non-toxicity and cytocompatibility with dental tissues and bone. The reality that nanocrystalline hydroxyapatite can be made of porous granules and scaffolds. Additionally, it has a massive loading potential indicating its use as a transporter for drugs or a regulated drug release mechanism in pharmaceutical research. This review aims to present existing nano-hydroxyapatite research developments as a drug carrier employed in bone tissue disorders locally and deliver poorly soluble drugs with reduced bioavailability. We have discussed the nano-hydroxyapatite role in the delivery of drugs (i.e. anti-resorptive, anti-cancer, and antibiotics), proteins, genetic material, and radionuclides.
Collapse
Affiliation(s)
- Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Sajal Salman
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Australia
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Naveed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Naveed Akhter Shad
- National Institute of Biotechnology and Genetic Engineering, Faisalabad, Pakistan
| | - Farooq Aziz
- Department of Physics, University of Sahiwal, Sahiwal, Pakistan
| |
Collapse
|
30
|
Bacteria-Based Microdevices for the Oral Delivery of Macromolecules. Pharmaceutics 2021; 13:pharmaceutics13101610. [PMID: 34683903 PMCID: PMC8537518 DOI: 10.3390/pharmaceutics13101610] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
The oral delivery of macromolecules is quite challenging due to environmental insults and biological barriers encountered along the gastrointestinal (GI) tract. Benefiting from their living characteristics, diverse bacterial species have been engineered as intelligent platforms to deliver various therapeutics. To tackle difficulties in oral delivery, innovative bacteria-based microdevices have been developed by virtue of advancements in synthetic biology and nanotechnology, with aims to overcome the instability and short half-life of macromolecules in the GI tract. In this review, we summarize the main classes of macromolecules that are produced and delivered through the oral ingestion of bacteria and bacterial derivatives. Furtherly, we discuss the engineering strategies and biomedical applications of these living microdevices in disease diagnosis, bioimaging, and treatment. Finally, we highlight the advantages as well as the limitations of these engineered bacteria used as platforms for the oral delivery of macromolecules and also propose their potential for clinical translation. The results summarized in this review article would contribute to the invention of next-generation bacteria-based systems for the oral delivery of macromolecules.
Collapse
|
31
|
A Dick T, Uludağ H. Mineralized polyplexes for gene delivery: Improvement of transfection efficiency as a consequence of calcium incubation and not mineralization. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112419. [PMID: 34579928 DOI: 10.1016/j.msec.2021.112419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 12/18/2022]
Abstract
Gene therapy is an emerging field in which nucleic acids are used to control protein expression. The necessity of delivering nucleic acids to specific cell types and intracellular sites demands the use of highly specialized gene carriers. As a carrier modification technique, mineralization has been successfully used to modify viral and non-viral carriers, providing new properties that ultimately aim to increase the transfection efficiency. However, for the specific case of polyplexes used in gene therapy, recent literature shows that interaction with calcium, a fundamental step of mineralization, might be effective to increase transfection efficiency, leaving an ambiguity about of the role of mineralization for this type of gene carriers. To answer this question and to reveal the properties responsible for increasing transfection efficiency, we mineralized poly(aspartic acid) coated polyplexes at various CaCl2 and Na3PO4 concentrations, and evaluated the resultant carriers for physicochemical and morphological characteristics, as well as transfection and delivery efficiency with MC3T3-E1 mouse osteoblastic cells. We found that both mineralization and calcium incubation positively affected the transfection efficiency and uptake of polyplexes in MC3T3-E1 cells. However, this effect originated from the properties achieved by polyplexes after the calcium incubation step that are maintained after mineralization, including particle size increase, improved pDNA binding, and adjustment of zeta potential. Considering that mineralization can be a longer process than calcium incubation, we find that calcium incubation might be sufficient and preferred if improved transfection efficiency in vitro is the only effect desired.
Collapse
Affiliation(s)
- Teo A Dick
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada.
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
32
|
Hu HJ, Liang X, Li HL, Wang HY, Gu JF, Sun LY, Xiao J, Hu JQ, Ni AM, Liu XY. Optimization of the Administration Strategy for the Armed Oncolytic Adenovirus ZD55-IL-24 in Both Immunocompromised and Immunocompetent Mouse Models. Hum Gene Ther 2021; 32:1481-1494. [PMID: 34155929 DOI: 10.1089/hum.2021.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
ZD55-IL-24 is an armed oncolytic adenovirus similar but superior to ONYX-015. Virotherapeutic strategies using ZD55-IL-24 have been demonstrated to be effective against several cancer types. However, it is unclear whether the traditional administration strategy is able to exert the maximal antitumor efficacy of ZD55-IL-24. In this study, we sought to optimize the administration strategy of ZD55-IL-24 in both A375-bearing immunocompromised mouse model and B16-bearing immunocompetent mouse model. Although the underlying antitumor mechanisms are quite different, the obtained results are similar in these two mouse tumor models. We find that the antitumor efficacy of ZD55-IL-24 increases as injection times increase in both of these two models. However, no obvious increase of efficacy is observed as the dose of each injection increases. Our further investigation reveals that the administration strategy of sustained ZD55-IL-24 therapy can achieve a better therapeutic effect than the traditional administration strategy of short-term ZD55-IL-24 therapy. Furthermore, there is no need to inject every day; every 2 or 3 days of injection achieves an equivalent therapeutic efficacy. Finally, we find that the sustained rather than the traditional short-term ZD55-IL-24 therapy can synergize with anti-PD-1 therapy to reject tumors in B16-bearing immunocompetent mouse model. These findings suggest that the past administration strategy of ZD55-IL-24 is in fact suboptimal and the antitumor efficacy can be further enhanced through administration strategy optimization. This study might shed some light on the development of clinically applicable administration regimens for ZD55-IL-24 therapy.
Collapse
Affiliation(s)
- Hai-Jun Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiu Liang
- School of Life Sciences and Technology, Tongji University, Shanghai, China; and
| | - Hai-Lang Li
- Department of Pharmacy, Xiamen Medical College, Xiamen, China
| | - Huai-Yuan Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jin-Fa Gu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Lan-Ying Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Jing Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jin-Qing Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ai-Min Ni
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xin-Yuan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
33
|
Mian-Ling Z, Yun-Qi C, Chao-Chun Z. Prader-Willi Syndrome: Molecular Mechanism and Epigenetic Therapy. Curr Gene Ther 2021; 20:36-43. [PMID: 32329685 DOI: 10.2174/1566523220666200424085336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/02/2020] [Accepted: 04/09/2020] [Indexed: 01/10/2023]
Abstract
Prader-Willi syndrome (PWS) is an imprinted neurodevelopmental disease characterized by cognitive impairments, developmental delay, hyperphagia, obesity, and sleep abnormalities. It is caused by a lack of expression of the paternally active genes in the PWS imprinting center on chromosome 15 (15q11.2-q13). Owing to the imprinted gene regulation, the same genes in the maternal chromosome, 15q11-q13, are intact in structure but repressed at the transcriptional level because of the epigenetic mechanism. The specific molecular defect underlying PWS provides an opportunity to explore epigenetic therapy to reactivate the expression of repressed PWS genes inherited from the maternal chromosome. The purpose of this review is to summarize the main advances in the molecular study of PWS and discuss current and future perspectives on the development of CRISPR/Cas9- mediated epigenome editing in the epigenetic therapy of PWS. Twelve studies on the molecular mechanism or epigenetic therapy of PWS were included in the review. Although our understanding of the molecular basis of PWS has changed fundamentally, there has been a little progress in the epigenetic therapy of PWS that targets its underlying genetic defects.
Collapse
Affiliation(s)
- Zhong Mian-Ling
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Chao Yun-Qi
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Zou Chao-Chun
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| |
Collapse
|
34
|
Gowsalya K, Yasothamani V, Vivek R. Emerging indocyanine green-integrated nanocarriers for multimodal cancer therapy: a review. NANOSCALE ADVANCES 2021; 3:3332-3352. [PMID: 36133722 PMCID: PMC9418715 DOI: 10.1039/d1na00059d] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/03/2021] [Indexed: 05/17/2023]
Abstract
Nanotechnology is a branch of science dealing with the development of new types of nanomaterials by several methods. In the biomedical field, nanotechnology is widely used in the form of nanotherapeutics. Therefore, the current biomedical research pays much attention to nanotechnology for the development of efficient cancer treatment. Indocyanine green (ICG) is a near-infrared tricarbocyanine dye approved by the Food and Drug Administration (FDA) for human clinical use. ICG is a biologically safe photosensitizer and it can kill tumor cells by producing singlet oxygen species and photothermal heat upon NIR irradiation. ICG has some limitations such as easy aggregation, rapid aqueous degradation, and a short half-life. To address these limitations, ICG is further formulated with nanoparticles. Therefore, ICG is integrated with organic nanomaterials (polymers, micelles, liposomes, dendrimers and protein), inorganic nanomaterials (magnetic, gold, mesoporous, calcium, and LDH based), and hybrid nanomaterials. The combination of ICG with nanomaterials provides highly efficient therapeutic effects. Nowadays, ICG is used for various biomedical applications, especially in cancer therapeutics. In this review, we mainly focus on ICG-based combined cancer nanotherapeutics for advanced cancer treatment.
Collapse
Affiliation(s)
- Karunanidhi Gowsalya
- Bio-Nano Therapeutics Research Laboratory, Cancer Research Program (CRP), School of Life Sciences, Department of Zoology, Bharathiar University Coimbatore-641 046 India
| | - Vellingiri Yasothamani
- Bio-Nano Therapeutics Research Laboratory, Cancer Research Program (CRP), School of Life Sciences, Department of Zoology, Bharathiar University Coimbatore-641 046 India
| | - Raju Vivek
- Bio-Nano Therapeutics Research Laboratory, Cancer Research Program (CRP), School of Life Sciences, Department of Zoology, Bharathiar University Coimbatore-641 046 India
| |
Collapse
|
35
|
Suo F, Pan M, Li Y, Yan Q, Hu H, Hou L. Mesenchymal Stem Cells Cultured in 3D System Inhibit Non-Small Cell Lung Cancer Cells through p38 MAPK and CXCR4/AKT Pathways by IL-24 Regulating. Mol Biol 2021. [DOI: 10.1134/s0026893321030110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Chen C, Shen M, Liao H, Guo Q, Fu H, Yu J, Duan Y. A paclitaxel and microRNA-124 coloaded stepped cleavable nanosystem against triple negative breast cancer. J Nanobiotechnology 2021; 19:55. [PMID: 33632232 PMCID: PMC7905927 DOI: 10.1186/s12951-021-00800-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background Triple negative breast cancer (TNBC) is one of the most biologically aggressive breast cancers and lacks effective treatment options, resulting in a poor prognosis. Therefore, studies aiming to explore new therapeutic strategies for advanced TNBC are urgently needed. According to recent studies, microRNA-124 (miR124) not only inhibits tumour growth but also increases the sensitivity of TNBC to paclitaxel (PTX), suggesting that a platform combining PTX and miR124 may be an advanced solution for TNBC. Results Herein, we constructed a stepped cleavable calcium phosphate composite lipid nanosystem (CaP/LNS) to codeliver PTX and miR124 (PTX/miR124-NP). PTX/miR124-NP exhibited superior tumor microenvironment responsive ability, in which the surface PEG layer was shed in the mildly acidic environment of tumor tissues and exposed oligomeric hyaluronic acid (o-HA) facilitated the cellular uptake of CaP/LNS by targeting the CD44 receptor on the surface of tumor cells. Inside tumour cells, o-HA detached from CaP/LNS due to the reduction of disulfide bonds by glutathione (GSH) and inhibited tumour metastasis. Then, PTX and miR124 were sequentially released from CaP/LNS and exerted synergistic antitumour effects by reversing the Epithelial-Mesenchymal Transition (EMT) process in MDA-MB-231 cells. Moreover, PTX/miR124-NP showed significant antitumour efficiency and excellent safety in mice bearing MDA-MB-231 tumours. Conclusion Based on these results, the codelivery of PTX and miR124 by the CaP/LNS nanosystem might be a promising therapeutic strategy for TNBC.![]()
Collapse
Affiliation(s)
- Chuanrong Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Ming Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China. .,NHC Key Laboratory of Reproduction Regulation, (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai, 200032, China.
| | - Hongze Liao
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
37
|
Hu HJ, Liang X, Li HL, Du CM, Hao JL, Wang HY, Gu JF, Ni AM, Sun LY, Xiao J, Hu JQ, Yuan H, Dai YS, Jin XT, Zhang KJ, Liu XY. The armed oncolytic adenovirus ZD55-IL-24 eradicates melanoma by turning the tumor cells from the self-state into the nonself-state besides direct killing. Cell Death Dis 2020; 11:1022. [PMID: 33257647 PMCID: PMC7705698 DOI: 10.1038/s41419-020-03223-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
ZD55-IL-24 is similar but superior to the oncolytic adenovirus ONYX-015, yet the exact mechanism underlying the observed therapeutic effect is still not well understood. Here we sought to elucidate the underlying antitumor mechanism of ZD55-IL-24 in both immunocompetent and immunocompromised mouse model. We find that ZD55-IL-24 eradicates established melanoma in B16-bearing immunocompetent mouse model not through the classic direct killing pathway, but mainly through the indirect pathway of inducing systemic antitumor immunity. Inconsistent with the current prevailing view, our further results suggest that ZD55-IL-24 can induce antitumor immunity in B16-bearing immunocompetent mouse model in fact not due to its ability to lyse tumor cells and release the essential elements, such as tumor-associated antigens (TAAs), but due to its ability to put a “nonself” label in tumor cells and then turn the tumor cells from the “self” state into the “nonself” state without tumor cell death. The observed anti-melanoma efficacy of ZD55-IL-24 in B16-bearing immunocompetent mouse model was practically caused only by the viral vector. In addition, we also notice that ZD55-IL-24 can inhibit tumor growth in B16-bearing immunocompetent mouse model through inhibiting angiogenesis, despite it plays only a minor role. In contrast to B16-bearing immunocompetent mouse model, ZD55-IL-24 eliminates established melanoma in A375-bearing immunocompromised mouse model mainly through the classic direct killing pathway, but not through the antitumor immunity pathway and anti-angiogenesis pathway. These findings let us know ZD55-IL-24 more comprehensive and profound, and provide a sounder theoretical foundation for its future modification and drug development.
Collapse
Affiliation(s)
- Hai-Jun Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xiu Liang
- School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China
| | - Hai-Lang Li
- Department of Pharmacy, Xiamen Medical College, 361023, Xiamen, China
| | - Chun-Ming Du
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Jia-Li Hao
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Huai-Yuan Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jin-Fa Gu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Ai-Min Ni
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Lan-Ying Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Jing Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jin-Qing Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Hao Yuan
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Yan-Song Dai
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Xiao-Ting Jin
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Kang-Jian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.
| | - Xin-Yuan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
38
|
Zhang Y, Dong Y, Fu H, Huang H, Wu Z, Zhao M, Yang X, Guo Q, Duan Y, Sun Y. Multifunctional tumor-targeted PLGA nanoparticles delivering Pt(IV)/siBIRC5 for US/MRI imaging and overcoming ovarian cancer resistance. Biomaterials 2020; 269:120478. [PMID: 33213862 DOI: 10.1016/j.biomaterials.2020.120478] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/08/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (Pt(II)) resistance is an important factor in the high mortality rates of ovarian cancer. Herein, we synthesized multifunctional tumor-targeted poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs-cRGD) for monitoring therapeutic effects by dual-mode imaging and overcoming cisplatin resistance. Uniformly sized NPs-cRGD demonstrated controlled and sustained release of drugs and genes, excellent gene loading and gene protection capacity, good storage stability and no serum-induced aggregation in vitro. NPs-cRGD demonstrated clear, targeting and prolonged ultrasound imaging and magnetic resonance imaging (MRI) in vivo. The targeting of NPs-cRGD combined with ultrasound facilitated nanoparticle penetrattion into cells; entry was time-dependent. NPs-cRGD escaped from lysosomes, thereby preventing siBIRC5 degradation, which enabled siBIRC5 to efficiently inhibit the antiapoptosis effects of BIRC5 in SKO3-DDP to overcome the antiapoptosis properties of resistant cells. Furthermore, Pt(IV) in NPs-cRGD exhausted glutathione (GSH), thereby increasing drug accumulation to effectively increase Pt(II) levels. The subsequent combination of Pt(II) with DNA prevented the expressions of genes and upregulated the expression of p53 to induce the mitochondria apoptosis pathway. The reduced GSH activity and the generation of Pt(II) further promoted high levels of reactive oxygen species (ROS) to induce cell apoptosis. Therefore, NPs-cRGD with ultrasound promoted the apoptosis of resistant ovarian cancer cells by multiple mechanisms, including increased cellular drug accumulation, reversed antiapoptotic effects by siBIRC5, and enhanced ROS levels. In a tumor-bearing nude mice model, NPs-cRGD with US demonstrated excellent tumor-targeting, high efficiency tumor inhibition and low systemic toxicity. Therefore, NPs-cRGD provides a means to monitor treatment processes and can be combined with ultrasound treatment to overcome ovarian cancer resistance in vitro and in vivo.
Collapse
Affiliation(s)
- Yanhua Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yang Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Hui Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Zhihua Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Meng Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Xupeng Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
39
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
40
|
Kong H, Zhao R, Zhang Q, Iqbal MZ, Lu J, Zhao Q, Luo D, Feng C, Zhang K, Liu X, Kong X. Biosilicified oncolytic adenovirus for cancer viral gene therapy. Biomater Sci 2020; 8:5317-5328. [PMID: 32779647 DOI: 10.1039/d0bm00681e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oncolytic adenoviruses (OAs) have shown great potential for cancer viral gene therapy in clinical studies. To date, clinical trials have shown that the curative efficacy of OAs is still limited by hepatic sequestration and preexisting neutralizing antibodies (nAbs), which decrease the accumulation of the OAs in tumors. Herein, with the biosilicification method, we encapsulated an OA encoding the anticancer gene Trail (OA-Trail) with silica, which significantly improved virus distribution and tumor inhibition. In vitro and in vivo results indicated that compared with the native OA, biosilicified OA-Trail (OA-Trail@SiO2) showed significantly reduced viral clearance in the liver and evaded nAb degradation, inducing an efficacious anticancer effect under the premise of biocompatibility. These achievements present an alternative strategy involving biosilicification for enhanced OA-based cancer gene therapy.
Collapse
Affiliation(s)
- Hao Kong
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cooper RC, Yang H. Duplex of Polyamidoamine Dendrimer/Custom-Designed Nuclear-Localization Sequence Peptide for Enhanced Gene Delivery. Bioelectricity 2020; 2:150-157. [PMID: 32856017 DOI: 10.1089/bioe.2020.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Dendrimers are an attractive alternative to viral vectors due to the low cost of production, larger genetic insert-carrying capacity, and added control over immune- and genotoxic complications through versatile functionalization. However, their transfection rates pale in comparison to their viral counterparts, resulting in widespread research efforts in the attempt to improve transfection efficiency. Materials and Methods: In this work, we designed a synthetic diblock nuclear-localization sequence peptide (NLS) (DDDDDDVKRKKKP) and complexed it with polyamidoamine (PAMAM) dendrimer G4 to form a duplex for gene delivery. We conducted transmission electron microscopy, gel mobility shift assay, and intracellular trafficking studies. We also assessed its transfection efficiency for the delivery of a green fluorescent protein-encoding plasmid (pGFP) to NIH3T3 cells. Results: PAMAM dendrimer G4, NLS, and plasmid DNA can form a stable three-part polyplex and gain enhanced entry into the nucleus. We found transfection efficiency, in large part, depends on the ratio of G4:NLS:plasmid. The triplex prepared at the ratio of 1:60:1 for G4:NLS:pGFP has been shown to be more significantly efficient in transfecting cells than the control group (G4/pGFP, 0.5:1). Conclusions: This new diblock NLS peptide can facilely complex with dendrimers to improve dendrimer-based gene transfection. It can also complex with other polycationic polymers to produce more potent nonviral duplex gene delivery vehicles.
Collapse
Affiliation(s)
- Remy C Cooper
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Hu Yang
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
42
|
Barry MA, Rubin JD, Lu SC. Retargeting adenoviruses for therapeutic applications and vaccines. FEBS Lett 2020; 594:1918-1946. [PMID: 31944286 PMCID: PMC7311308 DOI: 10.1002/1873-3468.13731] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022]
Abstract
Adenoviruses (Ads) are robust vectors for therapeutic applications and vaccines, but their use can be limited by differences in their in vitro and in vivo pharmacologies. This review emphasizes that there is not just one Ad, but a whole virome of diverse viruses that can be used as therapeutics. It discusses that true vector targeting involves not only retargeting viruses, but importantly also detargeting the viruses from off-target cells.
Collapse
Affiliation(s)
- Michael A Barry
- Department of Medicine, Division of Infectious Diseases, Department of Immunology, Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey D Rubin
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Shao-Chia Lu
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
43
|
Zhou H, Qiu X, Shen Z. [T 1-weighted magnetic resonance imaging contrast agents and their theranostic nanoprobes]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:427-444. [PMID: 32376585 DOI: 10.12122/j.issn.1673-4254.2020.03.24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Magnetic resonance imaging (MRI) is an important imaging modality for clinical disease diagnosis, and nearly 50% of clinical MRI examinations require contrast agents to enhance the diagnostic sensitivity. This review provides a summary of the major MRI contrast agents and their classification, and the advantages and limits of the commercially available MRI contrast agents, and elaborates on the exceedingly small magnetic iron oxide nanoparticles (ES-MIONs), dotted core-shell iron and gadolinium hybrid nanoparticles (FeGd-HN) and exceedingly small gadolinium oxide nanoparticles (ES-GON). These nanoparticles can greatly improve the efficiency of T1-weighted MRI due to their high r1 value and low r2/r1 ratio, and are expected to be translated into clinical contrast agents for T1-weighted MRI. The authors also review the diagnostic and therapeutic integration system that combines MRI contrast agents with various tumor therapies, such as MRI-guided ferroptosis therapy, radiosensitization therapy, and photothermal therapy, which allow efficient treatment as well as real-time monitoring of tumors and serve as potential cancer therapy strategies. The possible future research directions in the field of MRI-based multifunctional diagnostic and therapeutic formulations are also discussed.
Collapse
Affiliation(s)
- Huimin Zhou
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Xiaozhong Qiu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Zheyu Shen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
44
|
Hromic-Jahjefendic A, Lundstrom K. Viral Vector-Based Melanoma Gene Therapy. Biomedicines 2020; 8:E60. [PMID: 32187995 PMCID: PMC7148454 DOI: 10.3390/biomedicines8030060] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
Gene therapy applications of oncolytic viruses represent an attractive alternative for cancer treatment. A broad range of oncolytic viruses, including adenoviruses, adeno-associated viruses, alphaviruses, herpes simplex viruses, retroviruses, lentiviruses, rhabdoviruses, reoviruses, measles virus, Newcastle disease virus, picornaviruses and poxviruses, have been used in diverse preclinical and clinical studies for the treatment of various diseases, including colon, head-and-neck, prostate and breast cancer as well as squamous cell carcinoma and glioma. The majority of studies have focused on immunotherapy and several drugs based on viral vectors have been approved. However, gene therapy for malignant melanoma based on viral vectors has not been utilized to its full potential yet. This review represents a summary of the achievements of preclinical and clinical studies using viral vectors, with the focus on malignant melanoma.
Collapse
Affiliation(s)
- Altijana Hromic-Jahjefendic
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | | |
Collapse
|
45
|
Brugada-Vilà P, Cascante A, Lázaro MÁ, Castells-Sala C, Fornaguera C, Rovira-Rigau M, Albertazzi L, Borros S, Fillat C. Oligopeptide-modified poly(beta-amino ester)s-coated AdNuPARmE1A: Boosting the efficacy of intravenously administered therapeutic adenoviruses. Theranostics 2020; 10:2744-2758. [PMID: 32194832 PMCID: PMC7052890 DOI: 10.7150/thno.40902] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/23/2019] [Indexed: 01/09/2023] Open
Abstract
Oncolytic adenoviruses are used as agents for the treatment of cancer. However, their potential is limited due to the high seroprevalence of anti-adenovirus neutralizing antibodies (nAbs) within the population and the rapid liver sequestration when systemically administered. To overcome these challenges, we explored using nanoparticle formulation to boost the efficacy of systemic oncolytic adenovirus administration. Methods: Adenovirus were conjugated with PEGylated oligopeptide-modified poly(β-amino ester)s (OM-pBAEs). The resulting coated viral formulation was characterized in terms of surface charge, size, aggregation state and morphology and tested for anti-adenovirus nAbs evasion and activity in cancer cells. In vivo pharmacokinetics, biodistribution, tumor targeting, and immunogenicity studies were performed. The antitumor efficacy of the oncolytic adenovirus AdNuPARmE1A coated with OM-pBAEs (SAG101) in the presence of nAbs was evaluated in pancreatic ductal adenocarcinoma (PDAC) mouse models. Toxicity of the coated formulation was analyzed in vivo in immunocompetent mice. Results: OM-pBAEs conjugated to adenovirus and generated discrete nanoparticles with a neutral charge and an optimal size. The polymeric coating with the reporter AdGFPLuc (CPEG) showed enhanced transduction and evasion of antibody neutralization in vitro. Moreover, systemic intravenous administration of the formulation showed improved blood circulation and reduced liver sequestration, substantially avoiding activation of nAb production. OM-pBAEs coating of the oncolytic adenovirus AdNuPARmE1A (SAG101) improved its oncolytic activity in vitro and enhanced antitumor efficacy in PDAC mouse models. The coated formulation protected virions from neutralization by nAbs, as antitumor efficacy was preserved in their presence but was completely lost in mice that received the non-formulated AdNuPARmE1A. Finally, coated-AdNuPARmE1A showed reduced toxicity when high doses of the formulation were administered. Conclusions: The developed technology represents a promising improvement for future clinical cancer therapy using oncolytic adenoviruses.
Collapse
|
46
|
You Y, Huang K, Liu X, Pan X, Zhi J, He Q, Shi H, An Z, Ma X, Huang W. Hydrophilic Ultralong Organic Nanophosphors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906733. [PMID: 32003926 DOI: 10.1002/smll.201906733] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/02/2020] [Indexed: 06/10/2023]
Abstract
Ultralong organic phosphorescence (UOP), enabling of persistent luminescence after removal of external excitation light, shows great promise in biological applications such as bioimaging in virtue of antibackground fluorescence interference. Despite of good biocompatibility and outstanding phosphorescent properties, most current organic phosphors are hydrophobic with poor water solubility in the form of bulk crystal with large size, limiting their potential in the biological field. Here, a facile and versatile approach is provided to obtain nanoscale hydrophilic phosphorescent phosphors (HPPs) by physically loading ultralong organic phosphors into hollow mesoporous silica nanoparticles. The as-prepared HPPs can be well suspended in aqueous solution and effectively internalized by HeLa cells with very low cytotoxicity. Such HPPs are successfully applied for afterglow bioimaging in living nude mice with a very high signal-to-noise ratio up to 31. The current study not only provides a universal strategy to realize UOP in aqueous media but also demonstrates their great potential for biomedical purposes as an advanced imaging indicator with long-lived emission lifetime.
Collapse
Affiliation(s)
- Yongqiang You
- State Key Laboratory of Advanced Welding and Joining (Shenzhen), Harbin Institute of Technology (Shenzhen), Xili University Town, Shenzhen, 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen, 518055, China
| | - Kaiwei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Xiaojia Liu
- State Key Laboratory of Advanced Welding and Joining (Shenzhen), Harbin Institute of Technology (Shenzhen), Xili University Town, Shenzhen, 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen, 518055, China
| | - Xi Pan
- State Key Laboratory of Advanced Welding and Joining (Shenzhen), Harbin Institute of Technology (Shenzhen), Xili University Town, Shenzhen, 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen, 518055, China
| | - Jiahuan Zhi
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Qianjun He
- Guangdong Provincial Key Laboratory of Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xuyuan Road, Nanshan District, Shenzhen, 518055, China
| | - Huifang Shi
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Zhongfu An
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Xing Ma
- State Key Laboratory of Advanced Welding and Joining (Shenzhen), Harbin Institute of Technology (Shenzhen), Xili University Town, Shenzhen, 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen, 518055, China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
- Institute of Flexible Electronics (IFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| |
Collapse
|
47
|
Wu J, Wu H, Nakagawa S, Gao J. Virus-derived materials: bury the hatchet with old foes. Biomater Sci 2020; 8:1058-1072. [DOI: 10.1039/c9bm01383k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Viruses, with special architecture and unique biological nature, can be utilized for various biomedical applications.
Collapse
Affiliation(s)
- Jiahe Wu
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Honghui Wu
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Shinsaku Nakagawa
- Department of Pharmaceutics
- Graduate School of Pharmaceutical Sciences
- Osaka University
- Suita
- Japan
| | - Jianqing Gao
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| |
Collapse
|
48
|
Zhan X, Nie X, Gao F, Zhang C, You YZ, Yu Y. An NIR-activated polymeric nanoplatform with ROS- and temperature-sensitivity for combined photothermal therapy and chemotherapy of pancreatic cancer. Biomater Sci 2020; 8:5931-5940. [DOI: 10.1039/d0bm01324b] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A functional nanoplatform with ROS- and temperature-sensitivity was reported to achieve combined cancer treatment under NIR stimulation.
Collapse
Affiliation(s)
- Xiang Zhan
- AnHui Provincial Hospital
- Cheeloo College of Medicine
- Shandong University
- Jinan
- China
| | - Xuan Nie
- Hefei National Laboratory for Physical Sciences at the Microscale
- CAS Key Laboratory of Soft Matter Chemistry
- Department of Polymer Science and Engineering
- University of Science and Technology of China
- Hefei 230026
| | - Fan Gao
- Hefei National Laboratory for Physical Sciences at the Microscale
- CAS Key Laboratory of Soft Matter Chemistry
- Department of Polymer Science and Engineering
- University of Science and Technology of China
- Hefei 230026
| | - Cong Zhang
- Department of Gastroenterology
- AnHui Provincial Hospital
- Division of Life Sciences and Medicine
- University of Science and Technology of China
- Hefei
| | - Ye-Zi You
- Hefei National Laboratory for Physical Sciences at the Microscale
- CAS Key Laboratory of Soft Matter Chemistry
- Department of Polymer Science and Engineering
- University of Science and Technology of China
- Hefei 230026
| | - Yue Yu
- AnHui Provincial Hospital
- Cheeloo College of Medicine
- Shandong University
- Jinan
- China
| |
Collapse
|
49
|
Biointerfacial self-assembly generates lipid membrane coated bacteria for enhanced oral delivery and treatment. Nat Commun 2019; 10:5783. [PMID: 31857577 PMCID: PMC6923387 DOI: 10.1038/s41467-019-13727-9] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/22/2019] [Indexed: 02/08/2023] Open
Abstract
The gut microbiota represents a huge community of microorganisms that play essential roles in immune modulation and homeostasis maintenance. Microbiota transplantation is an important approach to prevent and treat disease as it can inhibit pathogen colonization and positively modulate bacterial composition. However, the development of oral bacterial therapeutics has been restricted by low bioavailability and limited retention in the gastrointestinal tract. Here, we report a simple yet highly efficient method to coat gut microbes via biointerfacial supramolecular self-assembly. Coating can be performed within 15 min by simply vortexing with biocompatible lipids. Bacteria coated with an extra self-assembled lipid membrane exhibit significantly improved survival against environmental assaults and almost unchanged viability and bioactivity. We demonstrate their enhanced efficacies in oral delivery and treatment using two murine models of colitis. We suggest that biointerfacial supramolecular self-assembly may provide a unique platform to generate advanced bacterial therapeutics for the treatment of various diseases. Oral microbiota delivery is an approach to treat and prevent disease but suffers from low retention and bioavailability. Here the authors report on a lipid coating to protect against environmental assault maintaining viability and bioactivity of the bacteria and demonstrate effective application in a colitis model.
Collapse
|
50
|
Huang LL, Li X, Zhang J, Zhao QR, Zhang MJ, Liu AA, Pang DW, Xie HY. MnCaCs-Biomineralized Oncolytic Virus for Bimodal Imaging-Guided and Synergistically Enhanced Anticancer Therapy. NANO LETTERS 2019; 19:8002-8009. [PMID: 31626554 DOI: 10.1021/acs.nanolett.9b03193] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Oncolytic adenovirus (OA) is an ideal candidate for clinical anticancer treatment, because it can specifically replicate in tumor cells with high titer. However, its systemic administration is still hindered, because of severely compromised antitumor efficacy. Herein, an engineered OA was innovatively developed by enwrapping OA with calcium and manganese carbonates (MnCaCs) biomineral shell, which could protect the virus from removal of the host immune system and prolong its in vivo circulation. Upon accumulating in tumor sites, MnCaCs readily dissolved under the acidic microenvironment, releasing Mn2+ that could convert endogenous H2O2 into oxygen (O2) and then enhance the duplication ability of OA, thus significantly increased the antitumor efficacy. Meanwhile, Mn2+ and the increased O2 individually endowed the T1 modal magnetic resonance imaging (MRI) and photoacoustic imaging (PAI) feasibility, providing real-time monitoring information for the therapy. This versatile engineered OA demonstrated its promise for visible and efficient oncolytic virotherapy by systemic administration.
Collapse
Affiliation(s)
- Li-Li Huang
- School of Life Science , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Xue Li
- School of Life Science , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - JinFeng Zhang
- School of Life Science , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Qian Ru Zhao
- School of Life Science , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Ming Jing Zhang
- School of Life Science , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - An-An Liu
- College of Chemistry , Nankai University , Tianjing 300071 , People's Republic of China
| | - Dai-Wen Pang
- College of Chemistry , Nankai University , Tianjing 300071 , People's Republic of China
| | - Hai-Yan Xie
- School of Life Science , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| |
Collapse
|