1
|
Li K, Guo B, Gu J, Ta N, Gu J, Yu H, Sun M, Han T. Emerging advances in drug delivery systems (DDSs) for optimizing cancer complications. Mater Today Bio 2025; 30:101375. [PMID: 39759851 PMCID: PMC11699619 DOI: 10.1016/j.mtbio.2024.101375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
The management and treatment of tumor complications pose continuous challenges due to the inherent complexity. However, the advent of drug delivery systems (DDSs) brings promising opportunities to address the tumor complications using innovative technological approaches. This review focuses on common oncological complications, including cancer thrombosis, malignant serous effusion, tumor-associated infections, cancer pain, and treatment-related complications. Emphasis was placed on the application and potential of DDSs in mitigating and treating these tumor complications, and we delved into the underlying mechanisms of common cancer-associated complications, discussed the limitations of conventional treatments, and outlined the current status and potential development of DDSs for various complications in this review. Moreover, we have discussed the existing challenges in DDSs research, underscoring the need for addressing issues related to biocompatibility and targeting of DDSs, optimizing drug delivery routes, and enhancing delivery efficiency and precision. In conclusion, DDSs offer promising avenues for treating cancer complications, offering the potential for the development of more effective and safer drug delivery strategies, thereby improving the quality of life and survival rates of cancer patients.
Collapse
Affiliation(s)
- Kerui Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Bei Guo
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Junmou Gu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Na Ta
- Department of Neurology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
| | - Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Hao Yu
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Zhang Z, Wang R, Chen L. Drug Delivery System Targeting Cancer-Associated Fibroblast for Improving Immunotherapy. Int J Nanomedicine 2025; 20:483-503. [PMID: 39816375 PMCID: PMC11734509 DOI: 10.2147/ijn.s500591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of non-malignant cells that play a crucial role in the tumor microenvironment, increasingly recognized as key contributors to cancer progression, metastasis, and treatment resistance. So, targeting CAFs has always been considered an important part of cancer immunotherapy. However, targeting CAFs to improve the efficacy of tumor therapy is currently a major challenge. Nanomaterials show their unique advantages in the whole process. At present, nanomaterials have achieved significant accomplishments in medical applications, particularly in the field of cancer-targeted therapy, showing enormous potential. It has been confirmed that nanomaterials can not only directly target CAFs, but also interact with the tumor microenvironment (TME) and immune cells to affect tumorigenesis. As for the cancer treatment, nanomaterials could enhance the therapeutic effect in many ways. Therefore, in this review, we first summarized the current understanding of the complex interactions between CAFs and TME, immune cells, and tumor cells. Next, we discussed common nanomaterials in modern medicine and their respective impacts on the TME, CAFs, and interactions with tumors. Finally, we focus on the application of nano drug delivery system targeting CAFs in cancer therapy.
Collapse
Affiliation(s)
- Zhongsong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Rong Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Long Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| |
Collapse
|
3
|
Zhang Y, Chen X, Hu B, Zou B, Xu Y. Advancements in nanomedicine delivery systems: unraveling immune regulation strategies for tumor immunotherapy. Nanomedicine (Lond) 2024; 19:1821-1840. [PMID: 39011582 PMCID: PMC11418288 DOI: 10.1080/17435889.2024.2374230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
This review highlights the significant role of nanodrug delivery systems (NDDS) in enhancing the efficacy of tumor immunotherapy. Focusing on the integration of NDDS with immune regulation strategies, it explores their transformative impacts on the tumor microenvironment and immune response dynamics. Key advancements include the optimization of drug delivery through NDDS, targeting mechanisms like immune checkpoint blockade and modulating the immunosuppressive tumor environment. Despite the progress, challenges such as limited clinical efficacy and complex manufacturing processes persist. The review emphasizes the need for further research to optimize these systems, potentially revolutionizing cancer treatment by improving delivery efficiency, reducing toxicity and overcoming immune resistance.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Xi Chen
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Binbin Hu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| |
Collapse
|
4
|
Pan Y, Cheng J, Zhu Y, Zhang J, Fan W, Chen X. Immunological nanomaterials to combat cancer metastasis. Chem Soc Rev 2024; 53:6399-6444. [PMID: 38745455 DOI: 10.1039/d2cs00968d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Metastasis causes greater than 90% of cancer-associated deaths, presenting huge challenges for detection and efficient treatment of cancer due to its high heterogeneity and widespread dissemination to various organs. Therefore, it is imperative to combat cancer metastasis, which is the key to achieving complete cancer eradication. Immunotherapy as a systemic approach has shown promising potential to combat metastasis. However, current clinical immunotherapies are not effective for all patients or all types of cancer metastases owing to insufficient immune responses. In recent years, immunological nanomaterials with intrinsic immunogenicity or immunomodulatory agents with efficient loading have been shown to enhance immune responses to eliminate metastasis. In this review, we would like to summarize various types of immunological nanomaterials against metastasis. Moreover, this review will summarize a series of immunological nanomaterial-mediated immunotherapy strategies to combat metastasis, including immunogenic cell death, regulation of chemokines and cytokines, improving the immunosuppressive tumour microenvironment, activation of the STING pathway, enhancing cytotoxic natural killer cell activity, enhancing antigen presentation of dendritic cells, and enhancing chimeric antigen receptor T cell therapy. Furthermore, the synergistic anti-metastasis strategies based on the combinational use of immunotherapy and other therapeutic modalities will also be introduced. In addition, the nanomaterial-mediated imaging techniques (e.g., optical imaging, magnetic resonance imaging, computed tomography, photoacoustic imaging, surface-enhanced Raman scattering, radionuclide imaging, etc.) for detecting metastasis and monitoring anti-metastasis efficacy are also summarized. Finally, the current challenges and future prospects of immunological nanomaterial-based anti-metastasis are also elucidated with the intention to accelerate its clinical translation.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Junjie Cheng
- Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian, China.
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| |
Collapse
|
5
|
Rahmat JN, Liu J, Chen T, Li Z, Zhang Y. Engineered biological nanoparticles as nanotherapeutics for tumor immunomodulation. Chem Soc Rev 2024; 53:5862-5903. [PMID: 38716589 DOI: 10.1039/d3cs00602f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Biological nanoparticles, or bionanoparticles, are small molecules manufactured in living systems with complex production and assembly machinery. The products of the assembly systems can be further engineered to generate functionalities for specific purposes. These bionanoparticles have demonstrated advantages such as immune system evasion, minimal toxicity, biocompatibility, and biological clearance. Hence, bionanoparticles are considered the new paradigm in nanoscience research for fabricating safe and effective nanoformulations for therapeutic purposes. Harnessing the power of the immune system to recognize and eradicate malignancies is a viable strategy to achieve better therapeutic outcomes with long-term protection from disease recurrence. However, cancerous tissues have evolved to become invisible to immune recognition and to transform the tumor microenvironment into an immunosuppressive dwelling, thwarting the immune defense systems and creating a hospitable atmosphere for cancer growth and progression. Thus, it is pertinent that efforts in fabricating nanoformulations for immunomodulation are mindful of the tumor-induced immune aberrations that could render cancer nanotherapy inoperable. This review systematically categorizes the immunosuppression mechanisms, the regulatory immunosuppressive cellular players, and critical suppressive molecules currently targeted as breakthrough therapies in the clinic. Finally, this review will summarize the engineering strategies for affording immune moderating functions to bionanoparticles that tip the tumor microenvironment (TME) balance toward cancer elimination, a field still in the nascent stage.
Collapse
Affiliation(s)
- Juwita N Rahmat
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117585, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yong Zhang
- Department of Biomedical Engineering, College of Engineering, The City University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
6
|
Pan H, Yang S, Gao L, Zhou J, Cheng W, Chen G, Shuhang W, Li N, Veranič P, Musiol R, Cai Q, Shubhra QT. At the crossroad of nanotechnology and cancer cell membrane coating: Expanding horizons with engineered nanoplatforms for advanced cancer therapy harnessing homologous tumor targeting. Coord Chem Rev 2024; 506:215712. [DOI: 10.1016/j.ccr.2024.215712] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Huang Q, Ge Y, He Y, Wu J, Tong Y, Shang H, Liu X, Ba X, Xia D, Peng E, Chen Z, Tang K. The Application of Nanoparticles Targeting Cancer-Associated Fibroblasts. Int J Nanomedicine 2024; 19:3333-3365. [PMID: 38617796 PMCID: PMC11012801 DOI: 10.2147/ijn.s447350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAF) are the most abundant stromal cells in the tumor microenvironment (TME), especially in solid tumors. It has been confirmed that it can not only interact with tumor cells to promote cancer progression and metastasis, but also affect the infiltration and function of immune cells to induce chemotherapy and immunotherapy resistance. So, targeting CAF has been considered an important method in cancer treatment. The rapid development of nanotechnology provides a good perspective to improve the efficiency of targeting CAF. At present, more and more researches have focused on the application of nanoparticles (NPs) in targeting CAF. These studies explored the effects of different types of NPs on CAF and the multifunctional nanomedicines that can eliminate CAF are able to enhance the EPR effect which facilitate the anti-tumor effect of themselves. There also exist amounts of studies focusing on using NPs to inhibit the activation and function of CAF to improve the therapeutic efficacy. The application of NPs targeting CAF needs to be based on an understanding of CAF biology. Therefore, in this review, we first summarized the latest progress of CAF biology, then discussed the types of CAF-targeting NPs and the main strategies in the current. The aim is to elucidate the application of NPs in targeting CAF and provide new insights for engineering nanomedicine to enhance immune response in cancer treatment.
Collapse
Affiliation(s)
- Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| |
Collapse
|
8
|
Zhou Z, Wang H, Li J, Jiang X, Li Z, Shen J. Recent progress, perspectives, and issues of engineered PD-L1 regulation nano-system to better cure tumor: A review. Int J Biol Macromol 2024; 254:127911. [PMID: 37939766 DOI: 10.1016/j.ijbiomac.2023.127911] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Currently, immune checkpoint blockade (ICB) therapies that target the programmed cell death ligand-1 (PD-L1) have been used as revolutionary cancer treatments in the clinic. Apart from restoring the antitumor response of cytotoxic T cells by blocking the interaction between PD-L1 on tumor cells and programmed cell death-1 (PD-1) on T cells, PD-L1 proteins were also newly revealed to possess the capacity to accelerate DNA damage repair (DDR) and enhance tumor growth through multiple mechanisms, leading to the impaired efficacy of tumor therapies. Nevertheless, current free anti-PD-1/PD-L1 therapy still suffered from poor therapeutic outcomes in most solid tumors due to the non-selective tumor accumulation, ineludible severe cytotoxic effects, as well as the common occurrence of immune resistance. Recently, nanoparticles with efficient tumor-targeting capacity, tumor-responsive prosperity, and versatility for combination therapy were identified as new avenues for PD-L1 targeting cancer immunotherapies. In this review, we first summarized the multiple functions of PD-L1 protein in promoting tumor growth, accelerating DDR, as well as depressing immunotherapy efficacy. Following this, the effects and mechanisms of current clinically widespread tumor therapies on tumor PD-L1 expression were discussed. Then, we reviewed the recent advances in nanoparticles for anti-PD-L1 therapy via using PD-L1 antibodies, small interfering RNA (siRNA), microRNA (miRNA), clustered, regularly interspaced, short palindromic repeats (CRISPR), peptide, and small molecular drugs. At last, we discussed the challenges and perspectives to promote the clinical application of nanoparticles-based PD-L1-targeting therapy.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jie Li
- College of Pharmacy, Wenzhou Medical University, Wenzhou 325000, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhangping Li
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| |
Collapse
|
9
|
Xiao Y, Wu M, Xue C, Wang Y. Recent Advances in the Development of Membrane-derived Vesicles for Cancer Immunotherapy. Curr Drug Deliv 2024; 21:403-420. [PMID: 37143265 DOI: 10.2174/1567201820666230504120841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023]
Abstract
The surface proteins on cell membranes enable the cells to have different properties, such as high biocompatibility, surface modifiability, and homologous targeting ability. Cell-membrane-derived vesicles have features identical to those of their parental cells, which makes them one of the most promising materials for drug delivery. Recently, as a result of the impressive effects of immunotherapy in cancer treatment, an increasing number of researchers have used cell-membrane-derived vesicles to enhance immune responses. To be more specific, the membrane vesicles derived from immune cells, tumor cells, bacteria, or engineered cells have the antigen presentation capacity and can trigger strong anti-tumor effects of the immune system. In this review, we first indicated a brief description of the vesicles and then introduced the detection technology and drug-loading methods for them. Secondly, we concluded the characteristics and applications of vesicles derived from different sources in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuai Xiao
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chunyu Xue
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuchong Wang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
10
|
Zhang J, Yang Y, Li K, Li J. Application of graphene oxide in tumor targeting and tumor therapy. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2551-2576. [PMID: 37768314 DOI: 10.1080/09205063.2023.2265171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023]
Abstract
Graphene oxide (GO), as a kind of two-dimensional sp2 carbon nanomaterials, has attracted great attention in many fields in the past decade. Due to its unique physical and chemical properties, GO is showing great promise in the field of biomedicine. For GO, all the atoms on its surface are exposed to the surface with ultra-high specific surface area, and a variety of groups on the surface, such as carboxyl, hydroxyl and epoxy groups, can effectively bind/load various biomolecules. Due to the availability of these groups, GO also possesses excellent hydrophilicity and biocompatibility for the modification of the desired biocompatible molecules or polymers on the surface of GO. The nano-network structure and hydrophobicity of GO enable it to load a large number of hydrophobic drugs containing benzene rings and it has been widely used as a multi-functional nano-carrier for chemotherapeutic drug or gene delivery. This review article will give an in-depth overview of the synthesis methods of GO, the advantages and disadvantages of GO used in nano-drug delivery system, the research progress of GO as a stimulus-responsive nano-drug carrier, and the application of these intelligent systems in cancer treatment.
Collapse
Affiliation(s)
- Jia Zhang
- College of Environmental & Chemical Engineering, Applied Chemistry Key Laboratory of Hebei Province, Key Laboratory of Nanobiotechnology of Hebei Province, Yanshan University, Qinhuangdao, Hebei Province, China
| | - Yibo Yang
- College of Environmental & Chemical Engineering, Applied Chemistry Key Laboratory of Hebei Province, Key Laboratory of Nanobiotechnology of Hebei Province, Yanshan University, Qinhuangdao, Hebei Province, China
| | - Kun Li
- College of Environmental & Chemical Engineering, Applied Chemistry Key Laboratory of Hebei Province, Key Laboratory of Nanobiotechnology of Hebei Province, Yanshan University, Qinhuangdao, Hebei Province, China
| | - Jian Li
- College of Environmental & Chemical Engineering, Applied Chemistry Key Laboratory of Hebei Province, Key Laboratory of Nanobiotechnology of Hebei Province, Yanshan University, Qinhuangdao, Hebei Province, China
| |
Collapse
|
11
|
Liu X, Xiao C, Xiao K. Engineered extracellular vesicles-like biomimetic nanoparticles as an emerging platform for targeted cancer therapy. J Nanobiotechnology 2023; 21:287. [PMID: 37608298 PMCID: PMC10463632 DOI: 10.1186/s12951-023-02064-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
Nanotechnology offers the possibility of revolutionizing cancer theranostics in the new era of precision oncology. Extracellular vesicles (EVs)-like biomimetic nanoparticles (EBPs) have recently emerged as a promising platform for targeted cancer drug delivery. Compared with conventional synthetic vehicles, EBPs have several advantages, such as lower immunogenicity, longer circulation time, and better targeting capability. Studies on EBPs as cancer therapeutics are rapidly progressing from in vitro experiments to in vivo animal models and early-stage clinical trials. Here, we describe engineering strategies to further improve EBPs as effective anticancer drug carriers, including genetic manipulation of original cells, fusion with synthetic nanomaterials, and direct modification of EVs. These engineering approaches can improve the anticancer performance of EBPs, especially in terms of tumor targeting effectiveness, stealth property, drug loading capacity, and integration with other therapeutic modalities. Finally, the current obstacles and future perspectives of engineered EBPs as the next-generation delivery platform for anticancer drugs are discussed.
Collapse
Affiliation(s)
- Xinyi Liu
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunxiu Xiao
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Xiao
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Tianfu Jingcheng Laboratory (Frontier Medical Center), Chengdu, 610041, China.
| |
Collapse
|
12
|
Sulaiman R, De P, Aske JC, Lin X, Dale A, Koirala N, Gaster K, Espaillat LR, Starks D, Dey N. Tumor-TME Bipartite Landscape of PD-1/PD-L1 in Endometrial Cancers. Int J Mol Sci 2023; 24:11079. [PMID: 37446260 DOI: 10.3390/ijms241311079] [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: 04/15/2023] [Revised: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
The bipartite landscape of tumor cells and stromal cells determines a tumor's response to treatment during disease management. In endometrial cancers (ECs), the mechanistic contribution of PD-L1/L2 and PD-1 signaling of the host's tumor microenvironment (TME) (CAF and immune cells) in the context of the tumor cells is elusive. To understand the tumor-stroma-immune crosstalk, we studied the compartmental pattern of PD-L1/L2 and PD-1 expression in EC tissues and their matched CAFs. Over 116 surgically resected tumors (T) and the tumor-adjacent normal tissues (N) were obtained from consented unselected consecutive patients. IHC was performed in T, N-epi-thelium, and the stromal mesenchymal environment (SME; mesenchyme) in the T and N tissues. The staining intensity and distribution patterns of PD-L1/L2 and PD-1 in the FFPE sections of T and N were evaluated by a pathologist using a standard scoring system of TPS and CPS. We tested the PD-L1/L2 and PD-1 immune landscape of tumor-TME pair and normal epithelial-stromal mesenchyme pairs from patients with different grades of disease vis-à-vis their CAF PD-L1 levels. We used qRT-PCR to determine the expressions of mRNAs, while the flow cytometry and ICC determined the level of expression of proteins. We observed higher levels of PD-L1 mRNA and protein expression in primary CAFs from the resected tumor tissue compared to the tumor-adjacent normal tissues. We also determined the expression of patients' soluble PD-L1/L2 as peripheral readouts of PD-L1/L2 and PD-1. As we evaluated the results in the context of their pathological parameters, such as grades, stages, lymphovascular invasion, percentage of myometrial invasion, and dMMR in patients, the dominance of PD-L1 expression in TME was positively correlated to the higher pathological grades of tumors, and its relationship with the dMMR. Since the neutralization of CD8-positive cytotoxic T-cells is PD-L1-dependent, our data indicate that irrespective of the PD-L1 positivity of tumor cells, the PD-L1-positive CAFs can play a critical role in bringing out an additional load of PD-L1 for an effective engagement of PD-1 within a tumor mass.
Collapse
Affiliation(s)
- Raed Sulaiman
- Department of Pathology, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Pradip De
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
- Department of Internal Medicine, University of South Dakota SSOM, Sioux Falls, SD 57108, USA
- Viecure, Greenwood Village, CO 80111, USA
| | - Jennifer C Aske
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Xiaoqian Lin
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Adam Dale
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Nischal Koirala
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Kris Gaster
- Assistant VP Outpatient Cancer Clinics, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Luis Rojas Espaillat
- Department of Gynecologic Oncology, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - David Starks
- Department of Gynecologic Oncology, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Nandini Dey
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
- Department of Internal Medicine, University of South Dakota SSOM, Sioux Falls, SD 57108, USA
| |
Collapse
|
13
|
Hu T, Huang Y, Liu J, Shen C, Wu F, He Z. Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy. Pharmaceutics 2023; 15:1821. [PMID: 37514008 PMCID: PMC10383408 DOI: 10.3390/pharmaceutics15071821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy can significantly prevent tumor growth and metastasis by activating the autoimmune system without destroying normal cells. Although cancer immunotherapy has made some achievements in clinical cancer treatment, it is still restricted by systemic immunotoxicity, immune cell dysfunction, cancer heterogeneity, and the immunosuppressive tumor microenvironment (ITME). Biomimetic cell-derived nanoparticles are attracting considerable interest due to their better biocompatibility and lower immunogenicity. Moreover, biomimetic cell-derived nanoparticles can achieve different preferred biological effects due to their inherent abundant source cell-relevant functions. This review summarizes the latest developments in biomimetic cell-derived nanoparticles for cancer immunotherapy, discusses the applications of each biomimetic system in cancer immunotherapy, and analyzes the challenges for clinical transformation.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuezhou Huang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chao Shen
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fengbo Wu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
14
|
Zheng Y, Guo W, Hu L, Xiao Z, Yang X, Cao Z, Cao J. Long Circulating Cancer Cell-Targeted Bionic Nanocarriers Enable Synergistic Combinatorial Therapy in Colon Cancer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:22843-22853. [PMID: 37133278 DOI: 10.1021/acsami.3c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cancer nanomedicine treatment aims to achieve highly specific targeting and localization to cancer cells. Coating of nanoparticles with cell membranes endows them with homologous cellular mimicry, enabling nanoparticles to acquire new functions and properties, including homologous targeting and long circulation in vivo, and can enhance internalization by homologous cancer cells. Herein, we fused a human-derived HCT116 colon cancer cell membrane (cM) with a red blood cell membrane (rM) to fabricate an erythrocyte-cancer cell hybrid membrane (hM). Oxaliplatin and chlorin e6 (Ce6) co-encapsulated reactive oxygen species-responsive nanoparticles (NPOC) were camouflaged by hM and obtained a hybrid biomimetic nanomedicine (denoted as hNPOC) for colon cancer therapy. hNPOC exhibited prolonged circulation time and recognized homologous targeting ability in vivo since both rM and HCT116 cM proteins were maintained on the hNPOC surface. hNPOC showed enhanced homologous cell uptake in vitro and considerable homologous self-localization in vivo, producing effective synergistic chemophotodynamic therapy efficacy under irradiation with a homologous HCT116 tumor compared to that with a heterologous tumor. Together, the biomimetic hNPOC nanoparticles showed prolonged blood circulation and preferential cancer cell-targeted function in vivo to provide a bioinspired strategy for chemophotodynamic synergistic therapy of colon cancer.
Collapse
Affiliation(s)
- Yunsheng Zheng
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, 510180 Guangzhou, P. R. China
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, 510180 Guangzhou, P. R. China
| | - Wenfeng Guo
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, 510180 Guangzhou, P. R. China
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, 510180 Guangzhou, P. R. China
| | - Lingwei Hu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, 511442 Guangzhou, P. R. China
| | - Zekai Xiao
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, 510006 Guangzhou, P. R. China
| | - Xianzhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, 511442 Guangzhou, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, 510006 Guangzhou, P. R. China
| | - Ziyang Cao
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, 510180 Guangzhou, P. R. China
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, 510180 Guangzhou, P. R. China
| | - Jie Cao
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, South China University of Technology, 510180 Guangzhou, P. R. China
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, 510180 Guangzhou, P. R. China
| |
Collapse
|
15
|
Liu N, Zhang R, Shi Q, Jiang H, Zhou Q. Intelligent delivery system targeting PD-1/PD-L1 pathway for cancer immunotherapy. Bioorg Chem 2023; 136:106550. [PMID: 37121105 DOI: 10.1016/j.bioorg.2023.106550] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 05/02/2023]
Abstract
The drugs targeting the PD-1/PD-L1 pathway have gained abundant clinical applications for cancer immunotherapy. However, only a part of patients benefit from such immunotherapy. Thus, brilliant novel tactic to increase the response rate of patients is on the agenda. Nanocarriers, particularly the rationally designed intelligent delivery systems with controllable therapeutic agent release ability and improved tumor targeting capacity, are firmly recommended. In light of this, state-of-the-art nanocarriers that are responsive to tumor-specific microenvironments (internal stimuli, including tumor acidic microenvironment, high level of GSH and ROS, specifically upregulated enzymes) or external stimuli (e.g., light, ultrasound, radiation) and release the target immunomodulators at tumor sites feature the advantages of increased anti-tumor potency but decreased off-target toxicity. Given the fantastic past achievements and the rapid developments in this field, the future is promising. In this review, intelligent delivery platforms targeting the PD-1/PD-L1 axis are attentively appraised. Specifically, mechanisms of the action of these stimuli-responsive drug release platforms are summarized to raise some guidelines for prior PD-1/PD-L1-based nanocarrier designs. Finally, the conclusion and outlook in intelligent delivery system targeting PD-1/PD-L1 pathway for cancer immunotherapy are outlined.
Collapse
Affiliation(s)
- Ning Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Qiang Shi
- Moji-Nano Technology Co. Ltd., Yantai 264006, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Qihui Zhou
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266071, China; Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin 300038, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
16
|
Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm (Beijing) 2023; 4:e187. [PMID: 36654533 PMCID: PMC9834710 DOI: 10.1002/mco2.187] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2023] Open
Abstract
Cancer is a complex disease associated with a combination of abnormal physiological process and exhibiting dysfunctions in multiple systems. To provide effective treatment and diagnosis for cancer, current treatment strategies simultaneously focus on various tumor targets. Based on the rapid development of nanotechnology, nanocarriers have been shown to exhibit excellent potential for cancer therapy. Compared with nanoparticles with single functions, multifunctional nanoparticles are believed to be more aggressive and potent in the context of tumor targeting. However, the development of multifunctional nanoparticles is not simply an upgraded version of the original function, but involves a sophisticated system with a proper backbone, optimized modification sites, simple preparation method, and efficient function integration. Despite this, many well-designed multifunctional nanoparticles with promising therapeutic potential have emerged recently. Here, to give a detailed understanding and analyzation of the currently developed multifunctional nanoparticles, their platform structures with organic or inorganic backbones were systemically generalized. We emphasized on the functionalization and modification strategies, which provide additional functions to the nanoparticle. We also discussed the application combination strategies that were involved in the development of nanoformulations with functional crosstalk. This review thus provides an overview of the construction strategies and application advances of multifunctional nanoparticles.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Xingmei Duan
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuan ProvinceChina
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
17
|
Qian Y, Wang J, Bu W, Zhu X, Zhang P, Zhu Y, Fan X, Wang C. Targeted implementation strategies of precise photodynamic therapy based on clinical and technical demands. Biomater Sci 2023; 11:704-718. [PMID: 36472233 DOI: 10.1039/d2bm01384c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
With the development of materials science, photodynamic-based treatments have gradually entered clinics. Photodynamic therapy is ideal for cancer treatment due to its non-invasive and spatiotemporal properties and is the first to be widely promoted in clinical practice. However, the shortcomings resulting from the gap between technical and clinical demands, such as phototoxicity, low tissue permeability, and tissue hypoxia, limit its wide applications. This article reviews the available data regarding the pharmacological and clinical factors affecting the efficacy of photodynamic therapy, such as photosensitizers and oxygen supply, disease diagnosis, and other aspects of photodynamic therapy. In addition, the synergistic treatment of photodynamic therapy with surgery and nanotechnology is also discussed, which is expected to provide inspiration for the design of photodynamic therapy strategies.
Collapse
Affiliation(s)
- Yun Qian
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Jialun Wang
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.
| | - Wenbo Bu
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Xiaoyan Zhu
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Ping Zhang
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Yun Zhu
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China. .,Department of Pharmacy, Nanjing Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.,Nanjing Medical Center for Clinical Pharmacy, Nanjing 210008, Jiangsu Province, China
| | - Xiaoli Fan
- Dermatologic Surgery Department, Institute of dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China.
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China.
| |
Collapse
|
18
|
Xu J, Cao W, Wang P, Liu H. Tumor-Derived Membrane Vesicles: A Promising Tool for Personalized Immunotherapy. Pharmaceuticals (Basel) 2022; 15:ph15070876. [PMID: 35890175 PMCID: PMC9318328 DOI: 10.3390/ph15070876] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor-derived membrane vesicles (TDMVs) are non-invasive, chemotactic, easily obtained characteristics and contain various tumor-borne substances, such as nucleic acid and proteins. The unique properties of tumor cells and membranes make them widely used in drug loading, membrane fusion and vaccines. In particular, personalized vectors prepared using the editable properties of cells can help in the design of personalized vaccines. This review focuses on recent research on TDMV technology and its application in personalized immunotherapy. We elucidate the strengths and challenges of TDMVs to promote their application from theory to clinical practice.
Collapse
Affiliation(s)
- Jiabin Xu
- School of Stomatology, Xuzhou Medical University, Xuzhou 221004, China; (J.X.); (P.W.)
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Wenqiang Cao
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Jinan University, Zhuhai 519000, China;
| | - Penglai Wang
- School of Stomatology, Xuzhou Medical University, Xuzhou 221004, China; (J.X.); (P.W.)
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Hong Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Jinan University, Zhuhai 519000, China;
- Correspondence:
| |
Collapse
|