1
|
Yao Y, Zhang J, Huang K, Peng Y, Cheng S, Liu S, Zhou T, Chen J, Li H, Zhao Y, Wang H. Engineered CAF-cancer cell hybrid membrane biomimetic dual-targeted integrated platform for multi-dimensional treatment of ovarian cancer. J Nanobiotechnology 2025; 23:83. [PMID: 39910555 PMCID: PMC11796236 DOI: 10.1186/s12951-025-03165-9] [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: 10/15/2024] [Accepted: 01/25/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND The efficacy of current therapies for ovarian cancer is limited due to the multilevel and complex tumor microenvironment (TME), which induces drug resistance and tumor progression in a single treatment regimen. Additionally, poor targeting and insufficient tissue penetration are important constraints in ovarian cancer treatment. RESULT We constructed PH20-overexpressing cancer-associated fibroblast (CAF)-cancer hybrid-cell membrane vesicles (PH20/CCM) for the dual-targeted delivery of carboplatin (CBP) and siRNA targeting p65 (sip65) loaded on the poly (dimethyl diallyl ammonium chloride) (PDDA)-modified MXene (PMXene), named PMXene@CBP-sip65 (PMCS). The nanoparticle PH20/CCM@PMCS could penetrate the extracellular matrix of tumor tissues and target both cancer cells and CAFs. After tumor cell internalization, these nanoparticles significantly inhibited cancer cell proliferation, generated reactive oxygen species, induced endoplasmic reticulum stress, and triggered immunogenic cell death. After CAF internalization, they inhibited pro-tumor factor release and activated immune effects, promoting immune system infiltration. In an experiment with ID8 homograft-carrying mice, PH20/CCM@PMCS significantly improved tumor inhibition and enhanced immune infiltration in tumor tissues. CONCLUSION These new therapeutic nanoparticles can simultaneously target tumor cells, CAFs, immune cells, and the extracellular matrix, thereby increasing treatment sensitivity and improving the TME. Therefore, these TME-regulating nanoparticles, combining specificity, efficiency, and effectiveness, provide new insights into ovarian cancer treatment.
Collapse
Affiliation(s)
- Yuwei Yao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Jiarui Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Kexin Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yingying Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Shuangshuang Cheng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Shuangge Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Ting Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Jinhua Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Haojia Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Clinical Research Center of Cancer Immunotherapy, Wuhan, Hubei, 430022, China.
| |
Collapse
|
2
|
Liang C, Ding X, Li X, Jiang X, Yang H, Yang H, Liu K, Hou L. In situ self-reassembling nanosystem enhances PD-L1 blockade for cancer immunotherapy. J Control Release 2025; 377:767-780. [PMID: 39631699 DOI: 10.1016/j.jconrel.2024.11.075] [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: 10/23/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Although immune checkpoint inhibitors (ICIs) have made great progress in cancer treatment, their off-tumor distribution, low affinity of traditional ICIs and insufficient T cells infiltration at tumor site limit immunotherapeutic efficacy. Herein, we engineer a highly specific and effective PD-L1 inhibitor (PEC) that modulates the level of binding sites with PD-L1. Specifically, PEC is a hybrid system composed of E. coli membrane expressing PD-L1 binding protein and cancer cell membrane. Notably, PEC can target the tumor site, produce oxygen in response to H2O2, rupture into membrane fragments, and reassemble to form vesicles retaining the PD-L1 binding protein. Through in situ fracture and reassembly, PEC transforms from a hybrid membrane to a single E. coli membrane, leading to the increased density of PD-L1 binding protein. Consequently, the reassembled vesicles can bind to more PD-L1 on tumor cells and induce its degradation in lysosomes. Furthermore, the cGAS-STING signaling activators HZD is encapsulated into PEC to promote T cells infiltration. We demonstrate that PEC@HZD achieves sequential T cells recruitment and functional enhancement, thus stimulating a powerful antitumor immune response. This work provides a new perspective on tailoring ICIs to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Chenglin Liang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Xinyi Ding
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Xinni Li
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaojuan Jiang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Heng Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Hanxiao Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Kaikai Liu
- Henan Key Laboratory of Diamond Optoelectronic Material and Devices, School of Physics and Laboratory of Zhongyuan Light, Zhengzhou University, Zhengzhou 450001, China.
| | - Lin Hou
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
3
|
Yan S, Gan Y, Xu H, Piao H. Bacterial carrier-mediated drug delivery systems: a promising strategy in cancer therapy. Front Bioeng Biotechnol 2025; 12:1526612. [PMID: 39845371 PMCID: PMC11750792 DOI: 10.3389/fbioe.2024.1526612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Cancer is a major killer threatening modern human health and a leading cause of death worldwide. Due to the heterogeneity and complexity of cancer, traditional treatments have limited effectiveness. To address this problem, an increasing number of researchers and medical professionals are working to develop new ways to treat cancer. Bacteria have chemotaxis that can target and colonize tumor tissue, as well as activate anti-tumor immune responses, which makes them ideal for biomedical applications. With the rapid development of nanomedicine and synthetic biology technologies, bacteria are extensively used as carriers for drug delivery to treat tumors, which holds the promise of overcoming the limitations of conventional cancer treatment regimens. This paper summarizes examples of anti-cancer drugs delivered by bacterial carriers, and their strengths and weaknesses. Further, we emphasize the promise of bacterial carrier delivery systems in clinical translation.
Collapse
Affiliation(s)
- Sizuo Yan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Yu Gan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Huizhe Xu
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
- Central Laboratory, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
4
|
Li G, Pu S, You L, Gao Y, Zhong Y, Zhao H, Fan D, Lu X. Innovative Strategies in Oncology: Bacterial Membrane Vesicle-Based Drug Delivery Systems for Cancer Diagnosis and Therapy. Pharmaceutics 2025; 17:58. [PMID: 39861706 PMCID: PMC11768367 DOI: 10.3390/pharmaceutics17010058] [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: 11/15/2024] [Revised: 12/18/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Outer membrane vesicles (OMVs) are double-layered structures of nanoscale lipids released by gram-negative bacteria. They have the same membrane composition and characteristics as primitive cells, which enables them to penetrate cells and tissues efficiently. These OMVs exhibit excellent membrane stability, immunogenicity, safety, and permeability (which makes it easier for them to penetrate into tumour tissue), making them suitable for developing cancer vaccines and drug delivery systems. Recent studies have focused on engineering OMVs to enhance tumour-targeting capabilities, reduce toxicity, and extend circulation time in vivo. This article reviews the latest progress in OMV engineering for tumour treatment and discusses the challenges associated with the use of OMV-based antitumour therapy in clinical practice.
Collapse
Affiliation(s)
- Guodong Li
- College of Life Sciences, Northwest University, Xi’an 710069, China; (G.L.)
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Shuangpeng Pu
- College of Life Sciences, Northwest University, Xi’an 710069, China; (G.L.)
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Lisiyao You
- College of Life Sciences, Northwest University, Xi’an 710069, China; (G.L.)
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Yuexia Zhong
- Outpatient Department of the Second Affiliated Hospital of the Fourth Military Medical University, Xi’an 710032, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China;
| | - Dong Fan
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China;
| | - Xiyan Lu
- Outpatient Department of the Second Affiliated Hospital of the Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
5
|
Aalhate M, Mahajan S, Dhuri A, Singh PK. Biohybrid nano-platforms manifesting effective cancer therapy: Fabrication, characterization, challenges and clinical perspective. Adv Colloid Interface Sci 2025; 335:103331. [PMID: 39522420 DOI: 10.1016/j.cis.2024.103331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Nanotechnology-based delivery systems have brought a paradigm shift in the management of cancer. However, the main obstacles to nanocarrier-based delivery are their limited circulation duration, excessive immune clearance, inefficiency in interacting effectively in a biological context and overcoming biological barriers. This demands effective engineering of nanocarriers to achieve maximum efficacy. Nanocarriers can be maneuvered with biological components to acquire biological identity for further regulating their biodistribution and cell-to-cell cross-talk. Thus, the integration of synthetic and biological components to deliver therapeutic cargo is called a biohybrid delivery system. These delivery systems possess the advantage of synthetic nanocarriers, such as high drug loading, engineerable surface, reproducibility, adequate communication and immune evasion ability of biological constituents. The biohybrid delivery vectors offer an excellent opportunity to harness the synergistic properties of the best entities of the two worlds for improved therapeutic outputs. The major spotlights of this review are different biological components, synthetic counterparts of biohybrid nanocarriers, recent advances in hybridization techniques, and the design of biohybrid delivery systems for cancer therapy. Moreover, this review provides an overview of biohybrid systems with therapeutic and diagnostic applications. In a nutshell, this article summarizes the advantages and limitations of various biohybrid nano-platforms, their clinical potential and future directions for successful translation in cancer management.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Anish Dhuri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India.
| |
Collapse
|
6
|
Xiang S, Khan A, Yao Q, Wang D. Recent advances in bacterial outer membrane vesicles: Effects on the immune system, mechanisms and their usage for tumor treatment. J Pharm Anal 2024; 14:101049. [PMID: 39840399 PMCID: PMC11750273 DOI: 10.1016/j.jpha.2024.101049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/27/2024] [Accepted: 07/19/2024] [Indexed: 01/23/2025] Open
Abstract
Tumor treatment remains a significant medical challenge, with many traditional therapies causing notable side effects. Recent research has led to the development of immunotherapy, which offers numerous advantages. Bacteria inherently possess motility, allowing them to preferentially colonize tumors and modulate the tumor immune microenvironment, thus influencing the efficacy of immunotherapy. Bacterial outer membrane vesicles (OMVs) secreted by gram-negative bacteria are nanoscale lipid bilayer structures rich in bacterial antigens, pathogen-associated molecular patterns (PAMPs), various proteins, and vesicle structures. These features allow OMVs to stimulate immune system activation, generate immune responses, and serve as efficient drug delivery vehicles. This dual capability enhances the effectiveness of immunotherapy combined with chemotherapy or phototherapy, thereby improving anticancer drug efficacy. Current research has concentrated on engineering OMVs to enhance production yield, minimize cytotoxicity, and improve the safety and efficacy of treatments. Consequently, OMVs hold great promise for applications in tumor immunotherapy, tumor vaccine development, and drug delivery. This article provides an overview of the structural composition and immune mechanisms of OMVs, details various OMVs modification strategies, and reviews the progress in using OMVs for tumor treatment and their anti-tumor mechanisms. Additionally, it discusses the challenges faced in translating OMV-based anti-tumor therapies into clinical practice, aiming to provide a comprehensive understanding of OMVs' potential for in-depth research and clinical application.
Collapse
Affiliation(s)
- Shuo Xiang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
- College of Advanced Materials Engineering, Jiaxing Nanhu University, 572 Yuexiu Road, Jiaxing, Zhejiang, 314001, China
| | - Arshad Khan
- Nanomedicine Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Qiufang Yao
- College of Advanced Materials Engineering, Jiaxing Nanhu University, 572 Yuexiu Road, Jiaxing, Zhejiang, 314001, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| |
Collapse
|
7
|
Liu C, Gao J, Cheng Y, Zhang S, Fu C. Homologous-adhering/targeting cell membrane- and cell-mediated delivery systems: a cancer-catch-cancer strategy in cancer therapy. Regen Biomater 2024; 12:rbae135. [PMID: 39811105 PMCID: PMC11729729 DOI: 10.1093/rb/rbae135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/09/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Low tumor enrichment remains a serious and urgent problem for drug delivery in cancer therapy. Accurate targeting of tumor sites is still a critical aim in cancer therapy. Though there have been a variety of delivery strategies to improve the tumor targeting and enrichment, biological barriers still cause most delivered guests to fail or be excreted before they work. Recently, cell membrane-based systems have attracted a huge amount of attention due to their advantages such as easy access, good biocompatibility and immune escape, which contribute to their biomimetic structures and specific surface proteins. Furthermore, cancer cell membrane-based delivery systems are referred to as homologous-targeting function in which they exhibit significantly high adhesion and internalization to homologous-type tumor sites or cells even though the exact mechanism is not entirely revealed. Here, we summarize the sources and characterizations of cancer cell membrane systems, including reconstructed single or hybrid membrane-based nano-/microcarriers, as well as engineered cancer cells. Additionally, advanced applications of these cancer cell membrane systems in cancer therapy are categorized and summarized according to the components of membranes. The potential factors related to homologous targeting of cancer cell membrane-based systems are also discussed. By discussing the applications, challenges and opportunities, we expect the cancer cell membrane-based homologous-targeting systems to have a far-reaching development in preclinic or clinics.
Collapse
Affiliation(s)
- Chenguang Liu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Jingjie Gao
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Yuying Cheng
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Shanshan Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Caiyun Fu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| |
Collapse
|
8
|
Jiang T, Zhan Y, Ding J, Song Z, Zhang Y, Li J, Su T. Biomimetic Cell Membrane-Coated Nanoparticles for Cancer Theranostics. ChemMedChem 2024; 19:e202400410. [PMID: 39264862 DOI: 10.1002/cmdc.202400410] [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: 05/28/2024] [Revised: 07/15/2024] [Indexed: 09/14/2024]
Abstract
Nanoparticles can enhance drugs accumulating at the tumor site and hold tremendous promise for achieving effective tumor treatment. However, due to the complexity of cancer heterogeneity and suppressive tumor microenvironment, the delivery of traditional nanoparticles has poor infiltration and off-target effects, making it difficult to control the drug release rate and causing off-target toxicity. In recent years, cell membrane-coated biomimetic nanoparticles have been developed, which have both the natural characteristics of biomembranes and the physical characteristics of traditional nanoparticles, thus improving the homologous targeting ability of nanoparticles to tumor cells and better biocompatibility. In this paper, we reviewed the application of single cell membrane and hybrid cell membrane-coated biomimetic nanoparticles in the integration for tumor diagnosis and treatment. We talked about the preparation methods of cell membrane-coated nanoparticles, the targeting mechanisms, and the effects of imaging and therapeutic outcomes of different cell membrane-coated biomimetic nanoparticles in detail. Finally, we discussed the existing problems and prospects of cell membrane-coated biomimetic nanomaterials.
Collapse
Affiliation(s)
- Tiantian Jiang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yiduo Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jiayao Ding
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Zheming Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yijing Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Ting Su
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
9
|
Singh A, Wu M, Ye TT, Brown AC, Wittenberg NJ. Engineering Planar Gram-Negative Outer Membrane Mimics Using Bacterial Outer Membrane Vesicles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:23289-23300. [PMID: 39453730 PMCID: PMC11542184 DOI: 10.1021/acs.langmuir.4c02632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 10/27/2024]
Abstract
Antibiotic resistance is a major challenge in modern medicine. The unique double membrane structure of Gram-negative bacteria limits the efficacy of many existing antibiotics and adds complexity to antibiotic development by limiting transport of antibiotics to the bacterial cytosol. New methods to mimic this barrier would enable high-throughput studies for antibiotic development. In this study, we introduce an innovative approach to modify outer membrane vesicles (OMVs) from Aggregatibacter actinomycetemcomitans, to generate planar supported lipid bilayer membranes. Our method first involves the incorporation of synthetic lipids into OMVs using a rapid freeze-thaw technique to form outer membrane hybrid vesicles (OM-Hybrids). Subsequently, these OM-Hybrids can spontaneously rupture when in contact with SiO2 surfaces to form a planar outer membrane supported bilayer (OM-SB). We assessed the formation of OM-Hybrids using dynamic light scattering and a fluorescence quenching assay. To analyze the formation of OM-SBs from OM-Hybrids we used quartz crystal microbalance with dissipation monitoring (QCM-D) and fluorescence recovery after photobleaching (FRAP). Additionally, we conducted assays to detect surface-associated DNA and proteins on OM-SBs. The interaction of an antimicrobial peptide, polymyxin B, with the OM-SBs was also assessed. These findings emphasize the capability of our platform to produce planar surfaces of bacterial outer membranes, which in turn, could function as a valuable tool for streamlining the development of antibiotics.
Collapse
Affiliation(s)
- Aarshi
N. Singh
- Department
of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Meishan Wu
- Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Tiffany T. Ye
- Department
of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Angela C. Brown
- Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Nathan J. Wittenberg
- Department
of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
10
|
Xu W, Maruyama S, Sato A, Niidome T. Bacterial membrane vesicles combined with nanoparticles for bacterial vaccines and cancer immunotherapy. Colloids Surf B Biointerfaces 2024; 243:114125. [PMID: 39079185 DOI: 10.1016/j.colsurfb.2024.114125] [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: 04/06/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/17/2024]
Abstract
Similar to mammalian cells, most bacteria can release nano-sized membrane vesicles (MVs) into the extracellular environment. MVs contain lipids, bioactive proteins, nucleic acids, and metabolites, and play important roles in microbial physiology. MVs have great potential for immunotherapeutic applications, such as bacterial vaccines and cancer immunotherapy. However, because of the diversity in content and heterogeneity in size of MVs, the clinical application of MVs has been limited. Recently, the use of MVs combined with nanoparticles (NPs) has been shown to be effective in improving the homogeneity, stability and function of MVs. In this review, we focus on studies of MVs combined with NPs (MV-NPs) and describe the use of these MV-NPs in biotechnology, especially in bacterial vaccine and cancer immunotherapy.
Collapse
Affiliation(s)
- Wei Xu
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan; International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan.
| | - Sayo Maruyama
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Akito Sato
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Takuro Niidome
- Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| |
Collapse
|
11
|
Khan M, Dong Y, Ullah R, Li M, Huang Q, Hu Y, Yang L, Luo Z. Recent Advances in Bacterium-Based Therapeutic Modalities for Melanoma Treatment. Adv Healthc Mater 2024; 13:e2401076. [PMID: 39375965 DOI: 10.1002/adhm.202401076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/16/2024] [Indexed: 10/09/2024]
Abstract
Melanoma is one of the most severe skin cancer indications with rapid progression and a high risk of metastasis. However, despite the accumulated advances in melanoma treatment including adjuvant radiation, chemotherapy, and immunotherapy, the overall melanoma treatment efficacy in the clinics is still not satisfactory. Interestingly, bacterial therapeutics have demonstrated unique properties for tumor-related therapeutic applications, such as tumor-targeted motility, tailorable cytotoxicity, and immunomodulatory capacity of the tumor microenvironment, which have emerged as a promising platform for melanoma therapy. Indeed, the recent advances in genetic engineering and nanotechnologies have boosted the application potential of bacterium-based therapeutics for treating melanoma by further enhancing their tumor-homing, cell-killing, drug delivery, and immunostimulatory capacities. This review provides a comprehensive summary of the state-of-the-art bacterium-based anti-melanoma modalities, which are categorized according to their unique functional merits, including tumor-specific cytotoxins, tumor-targeted drug delivery platforms, and immune-stimulatory agents. Furthermore, a perspective is provided discussing the potential challenges and breakthroughs in this area. The insights in this review may facilitate the development of more advanced bacterium-based therapeutic modalities for improved melanoma treatment efficacy.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yilong Dong
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325016, P. R. China
| | - Razi Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Lab for Vascular Implants College of Bioengineering Chongqing University, Chongqing, 400030, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Qiping Huang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| |
Collapse
|
12
|
Mengyuan H, Aixue L, Yongwei G, Qingqing C, Huanhuan C, Xiaoyan L, Jiyong L. Biomimetic nanocarriers in cancer therapy: based on intercellular and cell-tumor microenvironment communication. J Nanobiotechnology 2024; 22:604. [PMID: 39370518 PMCID: PMC11456251 DOI: 10.1186/s12951-024-02835-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Inspired by the concept of "natural camouflage," biomimetic drug delivery systems have emerged to address the limitations of traditional synthetic nanocarriers, such as poor targeting, susceptibility to identification and clearance, inadequate biocompatibility, low permeability, and systemic toxicity. Biomimetic nanocarriers retain the proteins, nucleic acids, and other components of the parent cells. They not only facilitate drug delivery but also serve as communication media to inhibit tumor cells. This paper delves into the communication mechanisms between various cell-derived biomimetic nanocarriers, tumor cells, and the tumor microenvironment, as well as their applications in drug delivery. In addition, the additional communication capabilities conferred on the modified biomimetic nanocarriers, such as targeting and environmental responsiveness, are outlined. Finally, we propose future development directions for biomimetic nanocarriers, hoping to inspire researchers in their design efforts and ultimately achieve clinical translation.
Collapse
Affiliation(s)
- He Mengyuan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Li Aixue
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Gu Yongwei
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Chai Qingqing
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Cai Huanhuan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Liu Xiaoyan
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, 200040, China.
| | - Liu Jiyong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Bai Z, Wang X, Liang T, Xu G, Cai J, Xu W, Yang K, Hu L, Pei P. Harnessing Bacterial Membrane Components for Tumor Vaccines: Strategies and Perspectives. Adv Healthc Mater 2024; 13:e2401615. [PMID: 38935934 DOI: 10.1002/adhm.202401615] [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: 05/01/2024] [Revised: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Tumor vaccines stand at the vanguard of tumor immunotherapy, demonstrating significant potential and promise in recent years. While tumor vaccines have achieved breakthroughs in the treatment of cancer, they still encounter numerous challenges, including improving the immunogenicity of vaccines and expanding the scope of vaccine application. As natural immune activators, bacterial components offer inherent advantages in tumor vaccines. Bacterial membrane components, with their safer profile, easy extraction, purification, and engineering, along with their diverse array of immune components, activate the immune system and improve tumor vaccine efficacy. This review systematically summarizes the mechanism of action and therapeutic effects of bacterial membranes and its derivatives (including bacterial membrane vesicles and hybrid membrane biomaterials) in tumor vaccines. Subsequently, the authors delve into the preparation and advantages of tumor vaccines based on bacterial membranes and hybrid membrane biomaterials. Following this, the immune effects of tumor vaccines based on bacterial outer membrane vesicles are elucidated, and their mechanisms are explained. Moreover, their advantages in tumor combination therapy are analyzed. Last, the challenges and trends in this field are discussed. This comprehensive analysis aims to offer a more informed reference and scientific foundation for the design and implementation of bacterial membrane-based tumor vaccines.
Collapse
Affiliation(s)
- Zhenxin Bai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuanyu Wang
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, People's Republic of China
| | - Tianming Liang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, P.R. China
| | - Guangyu Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jinzhou Cai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, P.R. China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Pei Pei
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, People's Republic of China
| |
Collapse
|
14
|
Wang KN, Li ZZ, Zhou K, Liu B, Rao L, Bu LL. Cell Membrane-Coated Nanoparticles for Dental, Oral, and Craniofacial Diseases. RESEARCH (WASHINGTON, D.C.) 2024; 7:0478. [PMID: 39296987 PMCID: PMC11409001 DOI: 10.34133/research.0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024]
Abstract
Dental, oral, and craniofacial diseases can substantially impact the quality of human life, thereby posing a serious public health concern. Although conventional therapies such as surgery have solved these problems largely, the prognosis of patients is not always satisfactory. Cell membrane-coated nanoparticles (CMCNPs) carry nanodrugs with the help of natural cell membranes, therefore utilizing their remarkable ability to interface and interact with their surrounding environment. These nanoparticles have demonstrated substantial advantages in drug targeting, prolonging blood circulation time, penetrating biofilms, and immune escape. With the assistance of CMCNPs, the therapeutic effects of dental, oral, and craniofacial diseases can reach a higher level. CMCNPs have been applied for dental, oral, and craniofacial diseases for various conditions such as head and neck cancer, periodontal disease, and oral biosignal detection. For the therapies of head and neck cancer, CMCNPs have been widely utilized as a tool of chemotherapy, phototherapy, and immunotherapy, while yet to be exploited in imaging technique. In the end, we summarized the challenges and prospectives of CMCNPs for dental, oral, and craniofacial diseases: large-scale production with uniform standards and high quantity, extensive application directions in dental, oral, and craniofacial regions (implant, endodontics), and the promotion of its clinical application.
Collapse
Affiliation(s)
- Kang-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral & Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral & Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
15
|
Luo Z, Cheng X, Feng B, Fan D, Liu X, Xie R, Luo T, Wegner SV, Ma D, Chen F, Zeng W. Engineering Versatile Bacteria-Derived Outer Membrane Vesicles: An Adaptable Platform for Advancing Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400049. [PMID: 38952055 PMCID: PMC11434149 DOI: 10.1002/advs.202400049] [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: 01/02/2024] [Revised: 06/13/2024] [Indexed: 07/03/2024]
Abstract
In recent years, cancer immunotherapy has undergone a transformative shift toward personalized and targeted therapeutic strategies. Bacteria-derived outer membrane vesicles (OMVs) have emerged as a promising and adaptable platform for cancer immunotherapy due to their unique properties, including natural immunogenicity and the ability to be engineered for specific therapeutic purposes. In this review, a comprehensive overview is provided of state-of-the-art techniques and methodologies employed in the engineering of versatile OMVs for cancer immunotherapy. Beginning by exploring the biogenesis and composition of OMVs, unveiling their intrinsic immunogenic properties for therapeutic appeal. Subsequently, innovative approaches employed to engineer OMVs are delved into, ranging from the genetic engineering of parent bacteria to the incorporation of functional molecules. The importance of rational design strategies is highlighted to enhance the immunogenicity and specificity of OMVs, allowing tailoring for diverse cancer types. Furthermore, insights into clinical studies and potential challenges utilizing OMVs as cancer vaccines or adjuvants are also provided, offering a comprehensive assessment of the current landscape and future prospects. Overall, this review provides valuable insights for researchers involved in the rapidly evolving field of cancer immunotherapy, offering a roadmap for harnessing the full potential of OMVs as a versatile and adaptable platform for cancer treatment.
Collapse
Affiliation(s)
- Ziheng Luo
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Xiang Cheng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Bin Feng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Duoyang Fan
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Xiaohui Liu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Ruyan Xie
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Ting Luo
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Seraphine V. Wegner
- Institute of Physiological Chemistry and PathobiochemistryUniversity of Münster48149MünsterGermany
| | - Dayou Ma
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Fei Chen
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic DiseasesChangsha410078China
| |
Collapse
|
16
|
Gholami A, Mohkam M, Soleimanian S, Sadraeian M, Lauto A. Bacterial nanotechnology as a paradigm in targeted cancer therapeutic delivery and immunotherapy. MICROSYSTEMS & NANOENGINEERING 2024; 10:113. [PMID: 39166136 PMCID: PMC11333603 DOI: 10.1038/s41378-024-00743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/02/2024] [Accepted: 06/23/2024] [Indexed: 08/22/2024]
Abstract
Cancer, a multifaceted and diverse ailment, presents formidable obstacles to traditional treatment modalities. Nanotechnology presents novel prospects for surmounting these challenges through its capacity to facilitate meticulous and regulated administration of therapeutic agents to malignant cells while concurrently modulating the immune system to combat neoplasms. Bacteria and their derivatives have emerged as highly versatile and multifunctional platforms for cancer nanotherapy within the realm of nanomaterials. This comprehensive review delves into the multifaceted and groundbreaking implementations of bacterial nanotechnology within cancer therapy. This review encompasses four primary facets: the utilization of bacteria as living conveyors of medicinal substances, the employment of bacterial components as agents that stimulate the immune system, the deployment of bacterial vectors as tools for delivering genetic material, and the development of bacteria-derived nano-drugs as intelligent nano-medications. Furthermore, we elucidate the merits and modalities of operation pertaining to these bacterial nano-systems, along with their capacity to synergize with other cutting-edge nanotechnologies, such as CRISPR-Cas systems. Additionally, we offer insightful viewpoints regarding the forthcoming trajectories and prospects within this expanding domain. It is our deduction that bacterial nanotechnology embodies a propitious and innovative paradigm in the realm of cancer therapy, which has the potential to provide numerous advantages and synergistic effects in enhancing the outcomes and quality of life for individuals afflicted with cancer.
Collapse
Affiliation(s)
- Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Mohkam
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Sadraeian
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Antonio Lauto
- School of Science, University of Western Sydney, Campbelltown, NSW 2560 Australia
- School of Medicine, University of Western Sydney, Campbelltown, NSW 2560 Australia
| |
Collapse
|
17
|
Singh AN, Wu M, Ye TT, Brown AC, Wittenberg NJ. Engineering Planar Gram-Negative Outer Membrane Mimics Using Bacterial Outer Membrane Vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.11.570829. [PMID: 39229024 PMCID: PMC11370475 DOI: 10.1101/2023.12.11.570829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Antibiotic resistance is a major challenge in modern medicine. The unique double membrane structure of gram-negative bacteria limits the efficacy of many existing antibiotics and adds complexity to antibiotic development by limiting transport of antibiotics to the bacterial cytosol. New methods to mimic this barrier would enable high-throughput studies for antibiotic development. In this study, we introduce an innovative approach to modify outer membrane vesicles (OMVs) from Aggregatibacter actinomycetemcomitans, to generate planar supported lipid bilayer membranes. Our method first involves the incorporation of synthetic lipids into OMVs using a rapid freeze-thaw technique to form outer membrane hybrid vesicles (OM-Hybrids). Subsequently, these OM-Hybrids can spontaneously rupture when in contact with SiO2 surfaces to form a planar outer membrane supported bilayer (OM-SB). We assessed the formation of OM-Hybrids using dynamic light scattering and a fluorescence quenching assay. To analyze the formation of OM-SBs from OM-Hybrids we used quartz crystal microbalance with dissipation monitoring (QCM-D) and fluorescence recovery after photobleaching (FRAP). Additionally, we conducted assays to detect surface-associated DNA and proteins on OM-SBs. The interaction of an antimicrobial peptide, polymyxin B, with the OM-SBs was also assessed. These findings emphasize the capability of our platform to produce planar surfaces of bacterial outer membranes, which in turn, could function as a valuable tool for streamlining the development of antibiotics.
Collapse
Affiliation(s)
- Aarshi N. Singh
- Department of Chemistry, Lehigh University, Bethlehem, PA, USA
| | - Meishan Wu
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA, USA
| | - Tiffany T. Ye
- Department of Chemistry, Lehigh University, Bethlehem, PA, USA
| | - Angela C. Brown
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA, USA
| | | |
Collapse
|
18
|
Meng Y, Kong C, Ma Y, Sun J, Zhang G. Bacterial outer membrane vesicles in the fight against cancer. Chin Med J (Engl) 2024:00029330-990000000-01174. [PMID: 39118214 PMCID: PMC11407815 DOI: 10.1097/cm9.0000000000003234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Indexed: 08/10/2024] Open
Abstract
ABSTRACT Bacterial outer membrane vesicles (OMVs) are diminutive vesicles naturally released by Gram-negative bacteria. These vesicles possess distinctive characteristics that attract attention for their potential use in drug administration and immunotherapy in cancer treatment. Therapeutic medicines may be delivered via OMVs directly to the tumor sites, thereby minimizing exposure to healthy cells and lowering the risk of systemic toxicity. Furthermore, the activation of the immune system by OMVs has been demonstrated to facilitate the recognition and elimination of cancer cells, which makes them a desirable tool for immunotherapy. They can also be genetically modified to carry specific antigens, immunomodulatory compounds, and small interfering RNAs, enhancing the immune response to cancerous cells and silencing genes associated with disease progression. Combining OMVs with other cancer treatments like chemotherapy and radiation has shown promising synergistic effects. This review highlights the crucial role of bacterial OMVs in cancer, emphasizing their potential as vectors for novel cancer targeted therapies. As researchers delve deeper into the complexities of these vesicles and their interactions with tumors, there is a growing sense of optimism that this avenue of study will bring positive outcomes and renewed hope to cancer patients in the foreseeable future.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Cuicui Kong
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yushu Ma
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| |
Collapse
|
19
|
Alimohammadvand S, Kaveh Zenjanab M, Mashinchian M, Shayegh J, Jahanban-Esfahlan R. Recent advances in biomimetic cell membrane-camouflaged nanoparticles for cancer therapy. Biomed Pharmacother 2024; 177:116951. [PMID: 38901207 DOI: 10.1016/j.biopha.2024.116951] [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: 04/14/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
The emerging strategy of biomimetic nanoparticles (NPs) via cellular membrane camouflage holds great promise in cancer therapy. This scholarly review explores the utilization of cellular membranes derived from diverse cellular entities; blood cells, immune cells, cancer cells, stem cells, and bacterial cells as examples of NP coatings. The camouflaging strategy endows NPs with nuanced tumor-targeting abilities such as self-recognition, homotypic targeting, and long-lasting circulation, thus also improving tumor therapy efficacy overall. The comprehensive examination encompasses a variety of cell membrane camouflaged NPs (CMCNPs), elucidating their underlying targeted therapy mechanisms and delineating diverse strategies for anti-cancer applications. Furthermore, the review systematically presents the synthesis of source materials and methodologies employed in order to construct and characterize these CMCNPs, with a specific emphasis on their use in cancer treatment.
Collapse
Affiliation(s)
- Sajjad Alimohammadvand
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Mashinchian
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Shayegh
- Department of Microbiology, Faculty of Veterinary and Agriculture, Islamic Azad University, Shabestar branch, Shabestar, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Gong N, Alameh MG, El-Mayta R, Xue L, Weissman D, Mitchell MJ. Enhancing in situ cancer vaccines using delivery technologies. Nat Rev Drug Discov 2024; 23:607-625. [PMID: 38951662 DOI: 10.1038/s41573-024-00974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/03/2024]
Abstract
In situ cancer vaccination refers to any approach that exploits tumour antigens available at a tumour site to induce tumour-specific adaptive immune responses. These approaches hold great promise for the treatment of many solid tumours, with numerous candidate drugs under preclinical or clinical evaluation and several products already approved. However, there are challenges in the development of effective in situ cancer vaccines. For example, inadequate release of tumour antigens from tumour cells limits antigen uptake by immune cells; insufficient antigen processing by antigen-presenting cells restricts the generation of antigen-specific T cell responses; and the suppressive immune microenvironment of the tumour leads to exhaustion and death of effector cells. Rationally designed delivery technologies such as lipid nanoparticles, hydrogels, scaffolds and polymeric nanoparticles are uniquely suited to overcome these challenges through the targeted delivery of therapeutics to tumour cells, immune cells or the extracellular matrix. Here, we discuss delivery technologies that have the potential to reduce various clinical barriers for in situ cancer vaccines. We also provide our perspective on this emerging field that lies at the interface of cancer vaccine biology and delivery technologies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, Center for BioAnalytical Chemistry, Hefei National Research Center for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Rakan El-Mayta
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Zhao C, Pan Y, Liu L, Zhang J, Wu X, Liu Y, Zhao XZ, Rao L. Hybrid Cellular Nanovesicles Block PD-L1 Signal and Repolarize M2 Macrophages for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311702. [PMID: 38456371 DOI: 10.1002/smll.202311702] [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: 12/15/2023] [Indexed: 03/09/2024]
Abstract
The PD1/PD-L1 immune checkpoint blocking is a promising therapy, while immunosuppressive tumor microenvironment (TME) and poor tumor penetration of therapeutic antibodies limit its efficacy. Repolarization of tumor-associated macrophages (TAMs) offers a potential method to ameliorate immunosuppression of TME and further boost T cell antitumor immunity. Herein, hybrid cell membrane biomimetic nanovesicles (hNVs) are developed by fusing M1 macrophage-derived nanovesicles (M1-NVs) and PD1-overexpressed tumor cell-derived nanovesicles (PD1-NVs) to improve cancer immunotherapy. The M1-NVs promote the transformation of M2-like TAMs to M1-like phenotype and further increase the release of pro-inflammatory cytokines, resulting in improved immunosuppressive TME. Concurrently, the PD1-NVs block PD1/PD-L1 pathway, which boosts cancer immunotherapy when combined with M1-NVs. In a breast cancer mouse model, the hNVs efficiently accumulate at the tumor site after intravenous injection and significantly inhibit the tumor growth. Mechanically, the M1 macrophages and CD8+ T lymphocytes in TME increase by twofold after the treatment, indicating effective immune activation. These results suggest the hNVs as a promising strategy to integrate TME improvement with PD1/PD-L1 blockade for cancer immunotherapy.
Collapse
Affiliation(s)
- Chenchen Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yuanwei Pan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Lujie Liu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jing Zhang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xianjia Wu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yu Liu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| |
Collapse
|
22
|
Das A, Sonar S, Kalele K, Subramaniyan V. Bacterial surface derived extracellular vesicles: A ground‐breaking approach in cancer therapy. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4. [DOI: 10.1002/ctd2.351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/14/2024] [Indexed: 11/04/2024]
Abstract
AbstractConventional cancer therapies are often limited by inherent and acquired drug resistance, non‐specific toxicity, and suboptimal drug delivery. Bacterial extracellular vesicles (BEVs), including outer membrane vesicles and membrane vesicles, present a novel therapeutic strategy for cancer treatment. BEVs were found in both Gram‐negative and Gram‐positive bacteria. This article provides a classified investigation of the therapeutic potential of BEVs in cancer treatment. We discuss the molecular mechanisms underlying their observed anti‐tumor effects, including the induction of apoptosis in cancer cells, suppression of angiogenesis within the tumor microenvironment, and stimulation of both innate and adaptive anti‐tumor immune responses. Moreover, BEVs being biocompatible opens avenue for targeted drug delivery systems, potentially improving the therapeutic index of conventional chemotherapeutics. Challenges to clinical translation, such as BEV heterogeneity and potential immunogenicity, are addressed. We also explore future directions for research and development, comprehensively highlighting how BEVs transform the landscape of precision oncology and cancer treatment, eventually improving patient outcomes.
Collapse
Affiliation(s)
- Asmit Das
- Department of Oral and Maxillofacial Pathology Neuron Institute of Applied Research Amravati India
| | - Swarup Sonar
- Center for Global Health Research Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences Chennai India
| | - Ketki Kalele
- Department of Oral and Maxillofacial Pathology Neuron Institute of Applied Research Amravati India
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences School of Medical and Life Sciences, Sunway University Sunway Malaysia
| |
Collapse
|
23
|
Zafar H, Zhang J, Raza F, Pan X, Hu Z, Feng H, Shen Q. Biomimetic gold nanocages incorporating copper-human serum albumin for tumor immunotherapy via cuproptosis-lactate regulation. J Control Release 2024; 372:446-466. [PMID: 38917953 DOI: 10.1016/j.jconrel.2024.06.059] [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: 03/28/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
Cancer immunotherapy remains a significant challenge due to insufficient proliferation of immune cells and the sturdy immunosuppressive tumor microenvironment. Herein, we proposed the hypothesis of cuproptosis-lactate regulation to provoke cuproptosis and enhance anti-tumor immunity. For this purpose, copper-human serum albumin nanocomplex loaded gold nanocages with bacterial membrane coating (BAu-CuNCs) were developed. The targeted delivery and disassembly of BAu-CuNCs in tumor cells initiated a cascade of reactions. Under near infrared (NIR) laser irradiation, the release of copper-human serum albumin (Cu-HSA) was enhanced that reacted with intratumoral glutathione (GSH) via a disulfide exchange reaction to liberate Cu2+ ions and exert cuproptosis. Subsequently, the cuproptosis effect triggered immunogenic cell death (ICD) in tumor by the release of damage associated molecular patterns (DAMPs) to realize anti-tumor immunity via robust production of cytotoxic T cells (CD8+) and helper T cells (CD4+). Meanwhile, under NIR irradiation, gold nanocages (AuNCs) promoted excessive reactive oxygen species (ROS) generation that played a primary role in inhibiting glycolysis, reducing the lactate and ATP level. The combine action of lower lactate level, ATP reduction and GSH depletion further sensitized the tumor cells to cuproptosis. Also, the lower lactate production led to the significant blockage of immunosuppressive T regulatory cells (Tregs) and boosted the anti-tumor immunity. Additionally, the effective inhibition of breast cancer metastasis to the lungs enhanced the anti-tumor therapeutic impact of BAu-CuNCs + NIR treatment. Hence, BAu-CuNCs + NIR concurrently induced cuproptosis, ICD and hindered lactate production, leading to the inhibition of tumor growth, remodeling of the immunosuppressive tumor microenvironment and suppression of lung metastasis. Therefore, leveraging cuproptosis-lactate regulation, this approach presents a novel strategy for enhanced tumor immunotherapy.
Collapse
Affiliation(s)
- Hajra Zafar
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jun Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Faisal Raza
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiuhua Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zongwei Hu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hanxiao Feng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China..
| |
Collapse
|
24
|
Peng X, Yang L, Yuan P, Ding X. Hybrid Cell Membrane-Based Nanoplatforms for Enhanced Immunotherapy against Cancer and Infectious Diseases. Adv Healthc Mater 2024; 13:e2304477. [PMID: 38709914 DOI: 10.1002/adhm.202304477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/20/2024] [Indexed: 05/08/2024]
Abstract
Immunotherapy based on nanoplatforms is a promising approach to treat cancer and infectious diseases, and it has achieved considerable progress in clinical practices. Cell membrane-based nanoplatforms endow nanoparticles with versatile characteristics, such as half-life extension, targeting ability, and immune-system regulation. However, monotypic cell membrane usually fails to provoke strong immune response for immunotherapy while maintaining good biosafety. The integration of different cell-membrane types provides a promising approach to construct multifunctional nanoplatforms for improved immunotherapeutic efficacy by enhancing immunogenicity or targeting function, evading immune clearance, or combining with other therapeutic modalities. In this review, the design principles, preparation strategies, and applications of hybrid cell membrane-based nanoplatforms for cancer and infection immunotherapy are first discussed. Furthermore, the challenges and prospects for the potential clinical translation of hybrid cell membrane-based nanoplatforms are discussed.
Collapse
Affiliation(s)
- Xinran Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Li Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xin Ding
- School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
25
|
Xu Z, Zhou H, Li T, Yi Q, Thakur A, Zhang K, Ma X, Qin JJ, Yan Y. Application of biomimetic nanovaccines in cancer immunotherapy: A useful strategy to help combat immunotherapy resistance. Drug Resist Updat 2024; 75:101098. [PMID: 38833804 DOI: 10.1016/j.drup.2024.101098] [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: 04/16/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024]
Abstract
Breakthroughs in actual clinical applications have begun through vaccine-based cancer immunotherapy, which uses the body's immune system, both humoral and cellular, to attack malignant cells and fight diseases. However, conventional vaccine approaches still face multiple challenges eliciting effective antigen-specific immune responses, resulting in immunotherapy resistance. In recent years, biomimetic nanovaccines have emerged as a promising alternative to conventional vaccine approaches by incorporating the natural structure of various biological entities, such as cells, viruses, and bacteria. Biomimetic nanovaccines offer the benefit of targeted antigen-presenting cell (APC) delivery, improved antigen/adjuvant loading, and biocompatibility, thereby improving the sensitivity of immunotherapy. This review presents a comprehensive overview of several kinds of biomimetic nanovaccines in anticancer immune response, including cell membrane-coated nanovaccines, self-assembling protein-based nanovaccines, extracellular vesicle-based nanovaccines, natural ligand-modified nanovaccines, artificial antigen-presenting cells-based nanovaccines and liposome-based nanovaccines. We also discuss the perspectives and challenges associated with the clinical translation of emerging biomimetic nanovaccine platforms for sensitizing cancer cells to immunotherapy.
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Haiyan Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tongfei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Qiaoli Yi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Abhimanyu Thakur
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Kui Zhang
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Xuelei Ma
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China.
| | - Jiang-Jiang Qin
- Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
26
|
Wang J, Liang S, Chen S, Ma T, Chen M, Niu C, Leng Y, Wang L. Bacterial outer membrane vesicle-cancer cell hybrid membrane-coated nanoparticles for sonodynamic therapy in the treatment of breast cancer bone metastasis. J Nanobiotechnology 2024; 22:328. [PMID: 38858780 PMCID: PMC11165797 DOI: 10.1186/s12951-024-02619-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
Breast cancer bone metastasis is a terminal-stage disease and is typically treated with radiotherapy and chemotherapy, which causes severe side effects and limited effectiveness. To improve this, Sonodynamic therapy may be a more safe and effective approach in the future. Bacterial outer membrane vesicles (OMV) have excellent immune-regulating properties, including modulating macrophage polarization, promoting DC cell maturation, and enhancing anti-tumor effects. Combining OMV with Sonodynamic therapy can result in synergetic anti-tumor effects. Therefore, we constructed multifunctional nanoparticles for treating breast cancer bone metastasis. We fused breast cancer cell membranes and bacterial outer membrane vesicles to form a hybrid membrane (HM) and then encapsulated IR780-loaded PLGA with HM to produce the nanoparticles, IR780@PLGA@HM, which had tumor targeting, immune regulating, and Sonodynamic abilities. Experiments showed that the IR780@PLGA@HM nanoparticles had good biocompatibility, effectively targeted to 4T1 tumors, promoted macrophage type I polarization and DC cells activation, strengthened anti-tumor inflammatory factors expression, and presented the ability to effectively kill tumors both in vitro and in vivo, which showed a promising therapeutic effect on breast cancer bone metastasis. Therefore, the nanoparticles we constructed provided a new strategy for effectively treating breast cancer bone metastasis.
Collapse
Affiliation(s)
- Jiahao Wang
- The School of Medicine, Nankai University, Tianjin, 300071, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shuailong Liang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Sijie Chen
- Department of Ultrasound Diagnosis, Second Xiangya Hospital, Central South University, Changsha, China
| | - Tianliang Ma
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mingyu Chen
- Department of Ultrasound Diagnosis, Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Leng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China.
| |
Collapse
|
27
|
Jeong Y, Han X, Vyas K, Irudayaraj J. Microbial β-Glucuronidase Hydrogel Beads Activate Chemotherapeutic Prodrug. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28093-28103. [PMID: 38775441 PMCID: PMC11164065 DOI: 10.1021/acsami.4c02568] [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: 02/14/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 06/07/2024]
Abstract
Bacteria-assisted chemotherapeutics have been highlighted as an alternative or supplementary approach to treating cancer. However, dynamic cancer-microbe studies at the in vitro level have remained a challenge to show the impact and effectiveness of microbial therapeutics due to the lack of relevant coculture models. Here, we demonstrate a hydrogel-based compartmentalized system for prodrug activation of a natural ingredient of licorice root, glycyrrhizin, by microbial β-glucuronidase (GUS). Hydrogel containment with Lactococcus lactis provides a favorable niche to encode GUS enzymes with excellent permeability and can serve as an independent ecosystem in the transformation of pro-apoptotic materials. Based on the confinement system of GUS expressing microbes, we quantitatively evaluated chemotherapeutic effects enhanced by microbial GUS enzyme in two dynamic coculture models in vitro (i.e., 2D monolayered cancer cells and 3D tumor spheroids). Our findings support the processes of prodrug conversion mediated by bacterial GUS enzyme which can enhance the therapeutic efficacy of a chemotherapy drug under dynamic coculture conditions. We expect our in vitro coculture platforms can be used for the evaluation of pharmacological properties and biological activity of xenobiotics as well as the potential impact of microbes on cancer therapeutics.
Collapse
Affiliation(s)
- Yoon Jeong
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, Urbana, Illinois 60801, United States
- Cancer
Center at Illinois, Carle-Illinois College
of Medicine, University of Illinois at Urbana−Champaign, Urbana, Illinois 60801, United States
- Biomedical
Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois 60801, United States
| | - Xiaoxue Han
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, Urbana, Illinois 60801, United States
- Cancer
Center at Illinois, Carle-Illinois College
of Medicine, University of Illinois at Urbana−Champaign, Urbana, Illinois 60801, United States
- Biomedical
Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois 60801, United States
| | - Khushali Vyas
- School
of Molecular and Cellular Biology, University
of Illinois at Urbana−Champaign, Urbana, Illinois 60801, United States
| | - Joseph Irudayaraj
- Department
of Bioengineering, University of Illinois
at Urbana−Champaign, Urbana, Illinois 60801, United States
- Cancer
Center at Illinois, Carle-Illinois College
of Medicine, University of Illinois at Urbana−Champaign, Urbana, Illinois 60801, United States
- Biomedical
Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois 60801, United States
- Carl
R. Woese Institute for Genomic Biology, Beckman Institute, Holonyak Micro and Nanotechnology Laboratory, Urbana, Illinois 60801, United States
| |
Collapse
|
28
|
Qureshi SA, Rafiya K, Awasthi S, Jain A, Nadaf A, Hasan N, Kesharwani P, Ahmad FJ. Biomembrane camouflaged nanoparticles: A paradigm shifts in targeted drug delivery system. Colloids Surf B Biointerfaces 2024; 238:113893. [PMID: 38631282 DOI: 10.1016/j.colsurfb.2024.113893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
Targeted drug delivery has emerged as a pivotal approach within precision medicine, aiming to optimize therapeutic efficacy while minimizing systemic side effects. Advanced biomimetic membrane-coated formulations have garnered significant interest from researchers as a promising strategy for targeted drug delivery, site-specific accumulation and heightened therapeutic outcomes. Biomimetic nanotechnology is able to retain the biological properties of the parent cell thus are able to exhibit superior targeting compared to conventional formulations. In this review, we have described different types of cell membrane camouflaged NPs. Mechanism of isolation and coating of the membranes along with the applications of each type of membrane and their mechanism to reach the desired site. Furthermore, a fusion of different membranes in order to prepare hybrid membrane biomimetic NPs which could possess better efficacy is discussed in detail in the review. Later, applications of the hybrid membrane-cloaked NPs along with current development were discussed in detail along with the challenges associated with it. Although membrane-cloaked NPs are currently in the preliminary stage of development, there is a huge potential to explore this biodegradable and biocompatible delivery system.
Collapse
Affiliation(s)
- Saba Asif Qureshi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Km Rafiya
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sakshi Awasthi
- Lloyd Institute of Management and Technology, Greater Noida, India
| | - Abhishek Jain
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Arif Nadaf
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
29
|
Yang M. Interaction between intestinal flora and gastric cancer in tumor microenvironment. Front Oncol 2024; 14:1402483. [PMID: 38835386 PMCID: PMC11148328 DOI: 10.3389/fonc.2024.1402483] [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: 03/17/2024] [Accepted: 05/01/2024] [Indexed: 06/06/2024] Open
Abstract
Gastric Cancer (GC) is a prevalent malignancy globally and is the third leading cause of cancer-related deaths. Recent researches focused on the correlation between intestinal flora and GC. Studies indicate that bacteria can influence the development of gastrointestinal tumors by releasing bacterial extracellular vesicles (BEVs). The Tumor microenvironment (TME) plays an important role in tumor survival, with the interaction between intestinal flora, BEVs, and TME directly impacting tumor progression. Moreover, recent studies have demonstrated that intestinal microflora and BEVs can modify TME to enhance the effectiveness of antitumor drugs. This review article provides an overview and comparison of the biological targets through which the intestinal microbiome regulates TME, laying the groundwork for potential applications in tumor diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Mingjin Yang
- Department of Gastrointestinal Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
30
|
Zhang M, Wang Y, Song Z, Lu Y, Zhao H, Wang Y, Lu P, Liu Y. Recent Progress of Bioinspired Cell Membrane in Cancer Immunotherapy. Clin Med Insights Oncol 2024; 18:11795549241236896. [PMID: 38645894 PMCID: PMC11032066 DOI: 10.1177/11795549241236896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/20/2024] [Indexed: 04/23/2024] Open
Abstract
By modifying immune cells, immunotherapy can activate immune response to establish long-term immune memory and prevent tumor recurrence. However, their effectiveness is largely constricted by the poor immunogenicity, immune escape, and immune tolerance of the tumor. This is related to the characteristics of the tumor itself, such as genome instability and mutation. The combination of various nanocarriers with tumor immunotherapy is beneficial for overcoming the shortcomings of traditional immunotherapy. Nanocarriers coated by cell membranes can extend blood circulation time, improve ability to evade immune clearance, and enhance targeting, thus significantly enhancing the efficacy of immunotherapy and showing great potential in tumor immunotherapy. This article reviews the application research progress of different types of cell membrane-modified nanocarriers in tumor immunotherapy, immunotherapy combination therapy, and tumor vaccines, and provides prospects for future research.
Collapse
Affiliation(s)
- Min Zhang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yuanhang Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Zhiyuan Song
- Department of Ultrasound Medicine, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Yimeng Lu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Houyu Zhao
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yihan Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Ping Lu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yanting Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| |
Collapse
|
31
|
Zhang S, Zhang X, Gao H, Zhang X, Sun L, Huang Y, Zhang J, Ding B. Cell Membrane-Coated Biomimetic Nanoparticles in Cancer Treatment. Pharmaceutics 2024; 16:531. [PMID: 38675192 PMCID: PMC11055162 DOI: 10.3390/pharmaceutics16040531] [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: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Nanoparticle-based drug delivery systems hold promise for cancer treatment by enhancing the solubility and stability of anti-tumor drugs. Nonetheless, the challenges of inadequate targeting and limited biocompatibility persist. In recent years, cell membrane nano-biomimetic drug delivery systems have emerged as a focal point of research and development, due to their exceptional traits, including precise targeting, low toxicity, and good biocompatibility. This review outlines the categorization and advantages of cell membrane bionic nano-delivery systems, provides an introduction to preparation methods, and assesses their applications in cancer treatment, including chemotherapy, gene therapy, immunotherapy, photodynamic therapy, photothermal therapy, and combination therapy. Notably, the review delves into the challenges in the application of various cell membrane bionic nano-delivery systems and identifies opportunities for future advancement. Embracing cell membrane-coated biomimetic nanoparticles presents a novel and unparalleled avenue for personalized tumor therapy.
Collapse
Affiliation(s)
- Shu Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 214122, China;
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Xiaojuan Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Huan Gao
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Xiaoqin Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Lidan Sun
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Yueyan Huang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Jie Zhang
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| | - Baoyue Ding
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China; (X.Z.); (H.G.); (X.Z.); (L.S.); (Y.H.)
| |
Collapse
|
32
|
Guo Q, Wang S, Xu R, Tang Y, Xia X. Cancer cell membrane-coated nanoparticles: a promising anti-tumor bionic platform. RSC Adv 2024; 14:10608-10637. [PMID: 38567339 PMCID: PMC10985588 DOI: 10.1039/d4ra01026d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Nanoparticle (NP) drug delivery systems have shown promise in tumor therapy. However, limitations such as susceptibility to immune clearance and poor targeting in a complex intercellular environment still exist. Recently, cancer cell membrane-encapsulated nanoparticles (CCM-NPs) constructed using biomimetic nanotechnology have been developed to overcome these problems. Proteins on the membrane surface of cancer cells can provide a wide range of activities for CCM-NPs, including immune escape and homologous cell recognition properties. Meanwhile, the surface of the cancer cell membrane exhibits obvious antigen enrichment, so that CCM-NPs can transmit tumor-specific antigen, activate a downstream immune response, and produce an effective anti-tumor effect. In this review, we first provided an overview of the functions of cancer cell membranes and summarized the preparation techniques and characterization methods of CCM-NPs. Then, we focused on the application of CCM-NPs in tumor therapy. In addition, we summarized the functional modifications of cancer cell membranes and compiled the patent applications related to CCM-NPs in recent years. Finally, we proposed the future challenges and directions of this technology in order to provide guidance for researchers in this field.
Collapse
Affiliation(s)
- Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Rubing Xu
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| | - Yingnan Tang
- School of Pharmacy, Hunan Vocational College of Science and Technology Changsha Hunan 410208 China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine Changsha Hunan 410208 China
| |
Collapse
|
33
|
Zheng P, He J, Fu Y, Yang Y, Li S, Duan B, Yang Y, Hu Y, Yang Z, Wang M, Liu Q, Zheng X, Hua L, Li W, Li D, Ding Y, Yang X, Bai H, Long Q, Huang W, Ma Y. Engineered Bacterial Biomimetic Vesicles Reprogram Tumor-Associated Macrophages and Remodel Tumor Microenvironment to Promote Innate and Adaptive Antitumor Immune Responses. ACS NANO 2024; 18:6863-6886. [PMID: 38386537 DOI: 10.1021/acsnano.3c06987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Tumor-associated macrophages (TAMs) are among the most abundant infiltrating leukocytes in the tumor microenvironment (TME). Reprogramming TAMs from protumor M2 to antitumor M1 phenotype is a promising strategy for remodeling the TME and promoting antitumor immunity; however, the development of an efficient strategy remains challenging. Here, a genetically modified bacterial biomimetic vesicle (BBV) with IFN-γ exposed on the surface in a nanoassembling membrane pore structure was constructed. The engineered IFN-γ BBV featured a nanoscale structure of protein and lipid vesicle, the existence of rich pattern-associated molecular patterns (PAMPs), and the costimulation of introduced IFN-γ molecules. In vitro, IFN-γ BBV reprogrammed M2 macrophages to M1, possibly through NF-κB and JAK-STAT signaling pathways, releasing nitric oxide (NO) and inflammatory cytokines IL-1β, IL-6, and TNF-α and increasing the expression of IL-12 and iNOS. In tumor-bearing mice, IFN-γ BBV demonstrated a targeted enrichment in tumors and successfully reprogrammed TAMs into the M1 phenotype; notably, the response of antigen-specific cytotoxic T lymphocyte (CTL) in TME was promoted while the immunosuppressive myeloid-derived suppressor cell (MDSC) was suppressed. The tumor growth was found to be significantly inhibited in both a TC-1 tumor and a CT26 tumor. It was indicated that the antitumor effects of IFN-γ BBV were macrophage-dependent. Further, the modulation of TME by IFN-γ BBV produced synergistic effects against tumor growth and metastasis with an immune checkpoint inhibitor in an orthotopic 4T1 breast cancer model which was insensitive to anti-PD-1 mAb alone. In conclusion, IFN-γ-modified BBV demonstrated a strong capability of efficiently targeting tumor and tuning a cold tumor hot through reprogramming TAMs, providing a potent approach for tumor immunotherapy.
Collapse
Affiliation(s)
- Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Ying Yang
- Cell Biology & Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, People's Republic of China
| | - Shuqin Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Kunming Medical University, Kunming 650500, People's Republic of China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Liangqun Hua
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Centers for Disease Control and Prevention, Kunming 530112, People's Republic of China
| | - Yiting Ding
- School of Life Sciences, Yunnan University, Kunming 650091, People's Republic of China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Hongmei Bai
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, People's Republic of China
| |
Collapse
|
34
|
Yu L, Zhou A, Jia J, Wang J, Ji X, Deng Y, Lin X, Wang F. Immunoactivity of a hybrid membrane biosurface on nanoparticles: enhancing interactions with dendritic cells to augment anti-tumor immune responses. Biomater Sci 2024; 12:1016-1030. [PMID: 38206081 DOI: 10.1039/d3bm01628e] [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: 01/12/2024]
Abstract
Nano-biointerfaces play a pivotal role in determining the functionality of engineered therapeutic nanoparticles, particularly in the context of designing nanovaccines to effectively activate immune cells for cancer immunotherapy. Unlike involving chemical reactions by conventional surface decorating strategies, cell membrane-coating technology offers a straightforward approach to endow nanoparticles with natural biosurfaces, enabling them to mimic and integrate into the intricate biosystems of the body to interact with specific cells under physiological conditions. In this study, cell membranes, in a hybrid formulation, derived from cancer and activated macrophage cells were found to enhance the interaction of nanoparticles (HMP) with dendritic cells (DCs) and T cells among the mixed immune cells from lymph nodes (LNs), which could be leveraged in the development of nanovaccines for anti-tumor therapy. After loading with an adjuvant (R837), the nanoparticles coated with a hybrid membrane (HMPR) demonstrated effectiveness in priming DCs both in vitro and in vivo, resulting in amplified anti-tumor immune responses compared to those of nanoparticles coated with a single type of membrane or those lacking a membrane coating. The elevated immunoactivity of nanoparticles achieved by incorporating a hybrid membrane biosurface provides us a more profound comprehension of the nano-immune interaction, which may significantly benefit the development of bioactive nanomaterials for advanced therapy.
Collapse
Affiliation(s)
- Luying Yu
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Ao Zhou
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Jingyan Jia
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Jieting Wang
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Xueyang Ji
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Yu Deng
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Xinhua Lin
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Fang Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
35
|
Fernández-Borbolla A, García-Hevia L, Fanarraga ML. Cell Membrane-Coated Nanoparticles for Precision Medicine: A Comprehensive Review of Coating Techniques for Tissue-Specific Therapeutics. Int J Mol Sci 2024; 25:2071. [PMID: 38396747 PMCID: PMC10889273 DOI: 10.3390/ijms25042071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Nanoencapsulation has become a recent advancement in drug delivery, enhancing stability, bioavailability, and enabling controlled, targeted substance delivery to specific cells or tissues. However, traditional nanoparticle delivery faces challenges such as a short circulation time and immune recognition. To tackle these issues, cell membrane-coated nanoparticles have been suggested as a practical alternative. The production process involves three main stages: cell lysis and membrane fragmentation, membrane isolation, and nanoparticle coating. Cell membranes are typically fragmented using hypotonic lysis with homogenization or sonication. Subsequent membrane fragments are isolated through multiple centrifugation steps. Coating nanoparticles can be achieved through extrusion, sonication, or a combination of both methods. Notably, this analysis reveals the absence of a universally applicable method for nanoparticle coating, as the three stages differ significantly in their procedures. This review explores current developments and approaches to cell membrane-coated nanoparticles, highlighting their potential as an effective alternative for targeted drug delivery and various therapeutic applications.
Collapse
Affiliation(s)
- Andrés Fernández-Borbolla
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Lorena García-Hevia
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Mónica L. Fanarraga
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| |
Collapse
|
36
|
Ruan Y, Zhuang H, Zeng X, Lin L, Wang X, Xue P, Xu S, Chen Q, Yan S, Huang W. Engineered Microbial Nanohybrids for Tumor-Mediated NIR II Photothermal Enhanced Ferroptosis/Cuproptosis and Immunotherapy. Adv Healthc Mater 2024; 13:e2302537. [PMID: 37742322 DOI: 10.1002/adhm.202302537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/20/2023] [Indexed: 09/26/2023]
Abstract
The colon tumor microenvironment has a high concentration of H2 S and glutathione, which is highly immunosuppressive and adverse to multiple therapeutic methodologies such as ferroptosis. Here, an engineered microbial nanohybrid based on Escherichia coli (E. coli) and Cu2 O nanoparticles to specific colon tumor therapy and immunosuppression reversion is reported. The as-prepared E. coli@Cu2 O hybrid can accumulate in tumor sites upon intravenous injection, and Cu2 O nanoparticles convert to Cux S by consuming the endogenous H2 S, which exhibits strong photothermal conversion at near-infrared II (NIR II) biological window. Furthermore, E. coli@Cu2 O is able to induce cellular ferroptosis and cuproptosis through inactivation of glutathione peroxidase 4 and aggregation of dihydrolipoamide S-acetyltransferase, respectively. Photothermal-enhanced ferroptosis/cuproptosis achieved by E. coli@Cu2 O reverses the immunosuppression of colon tumors by triggering dendritic cell maturation (about 30%) and T cell activation (about 50% CD8+ T cells). Concerted with immune checkpoint blockade, the engineered microbial nanohybrid can inhibit the growth of abscopal tumors upon NIR illumination. Overall, the designed microbial nanohybrid can achieve tumor-specific photothermal-enhanced ferroptosis/cuproptosis and immunosuppression reversion, showing promise in precise tumor therapy in future clinical translation.
Collapse
Affiliation(s)
- Yihang Ruan
- The Straits Laboratory of Flexible Electronics (SLoFE), Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
- Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fujian, 350117, China
| | - Huilan Zhuang
- The Straits Laboratory of Flexible Electronics (SLoFE), Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Xuemei Zeng
- Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fujian, 350117, China
| | - Lili Lin
- Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fujian, 350117, China
| | - Xuechun Wang
- Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350117, China
| | - Panpan Xue
- The Straits Laboratory of Flexible Electronics (SLoFE), Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Shan Xu
- Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fujian, 350117, China
| | - Qi Chen
- Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fujian, 350117, China
| | - Shuangqian Yan
- The Straits Laboratory of Flexible Electronics (SLoFE), Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics (FSCFE), MIIT Key Laboratory of Flexible Electronics (KLoFE), Northwestern Polytechnical University, Xi'an, Shanxi, 710072, China
- Key Laboratory of Flexible Electronics (KLoFE) & Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, Jiangsu, 211816, China
| |
Collapse
|
37
|
Lu Y, Fan L, Wang J, Hu M, Wei B, Shi P, Li J, Feng J, Zheng Y. Cancer Cell Membrane-Based Materials for Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306540. [PMID: 37814370 DOI: 10.1002/smll.202306540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/18/2023] [Indexed: 10/11/2023]
Abstract
The nanodelivery system provides a novel direction for disease diagnosis and treatment; however, its delivery effectiveness is restricted by the short biological half-life and inadequate tumor targeting. The immune evasion properties and homologous targeting capabilities of natural cell membranes, particularly those of cancer cell membranes (CCM), have gained significant interest. The integration of CCM and nanoparticles has resulted in the emergence of CCM-based nanoplatforms (CCM-NPs), which have gained significant attention due to their unique properties. CCM-NPs not only prolong the blood circulation time of core nanoparticles, but also direct them for homologous tumor targeting. Herein, the history and development of CCM-NPs as well as how these platforms have been used for biomedical applications are discussed. The application of CCM-NPs for cancer therapy will be described in detail. Translational efforts are currently under way and further research to address key areas of need will ultimately be required to facilitate the successful clinical adoption of CCM-NPs.
Collapse
Affiliation(s)
- Yongping Lu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
- Guangyuan Key Laboratory of Multifunctional Medical Hydrogel, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Linming Fan
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jun Wang
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Mingxiang Hu
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Baogang Wei
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Ping Shi
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Jinyan Feng
- Science and Technologv Innovation Center, Guangyuan Central Hospital, Guangyuan, 628000, China
| | - Yu Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
38
|
Han X, Gong C, Yang Q, Zheng K, Wang Z, Zhang W. Biomimetic Nano-Drug Delivery System: An Emerging Platform for Promoting Tumor Treatment. Int J Nanomedicine 2024; 19:571-608. [PMID: 38260239 PMCID: PMC10802790 DOI: 10.2147/ijn.s442877] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
With the development of nanotechnology, nanoparticles (NPs) have shown broad prospects as drug delivery vehicles. However, they exhibit certain limitations, including low biocompatibility, poor physiological stability, rapid clearance from the body, and nonspecific targeting, which have hampered their clinical application. Therefore, the development of novel drug delivery systems with improved biocompatibility and high target specificity remains a major challenge. In recent years, biofilm mediated biomimetic nano-drug delivery system (BNDDS) has become a research hotspot focus in the field of life sciences. This new biomimetic platform uses bio-nanotechnology to encapsulate synthetic NPswithin biomimetic membrane, organically integrating the low immunogenicity, low toxicity, high tumor targeting, good biocompatibility of the biofilm with the adjustability and versatility of the nanocarrier, and shows promising applications in the field of precision tumor therapy. In this review, we systematically summarize the new progress in BNDDS used for optimizing drug delivery, providing a theoretical reference for optimizing drug delivery and designing safe and efficient treatment strategies to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Xiujuan Han
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Qingru Yang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Kaile Zheng
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
| | - Zhuo Wang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Wei Zhang
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
39
|
Li S, Meng X, Peng B, Huang J, Liu J, Xiao H, Ma L, Liu Y, Tang J. Cell membrane-based biomimetic technology for cancer phototherapy: Mechanisms, recent advances and perspectives. Acta Biomater 2024; 174:26-48. [PMID: 38008198 DOI: 10.1016/j.actbio.2023.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/04/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Despite significant advances in medical technology and antitumour treatments, the diagnosis and treatment of tumours have undergone remarkable transformations. Noninvasive phototherapy methods, such as photodynamic therapy (PDT) and photothermal therapy (PTT), have gained significant interest in antitumour medicine. However, traditional photosensitisers or photothermal agents face challenges like immune system recognition, rapid clearance from the bloodstream, limited tumour accumulation, and phototoxicity concerns. Researchers combine photosensitisers or photothermal agents with natural cell membranes to overcome these obstacles to create a nano biomimetic therapeutic platform. When used to coat nanoparticles, red blood cells, platelets, cancer cells, macrophages, lymphocytes, and bacterial outer membranes could provide prolonged circulation, tumour targeting, immune stimulation, or antigenicity. This article covers the principles of cellular membrane biomimetic nanotechnology and phototherapy, along with recent advancements in applying nano biomimetic technology to PDT, PTT, PCT, and combined diagnosis and treatment. Furthermore, the challenges and issues of using nano biomimetic nanoparticles in phototherapy are discussed. STATEMENT OF SIGNIFICANCE: Currently, there has been significant progress in the field of cell membrane biomimetic technology. Researchers are exploring its potential application in tumor diagnosis and treatment through phototherapy. Scholars have conducted extensive research on combining cell membrane technology and phototherapy in anticancer diagnosis and treatment. This review aims to highlight the mechanisms of phototherapy and the latest advancements in single phototherapy (PTT, PDT) and combination phototherapy (PCT, PRT, and PIT), as well as diagnostic approaches. The review provides an overview of various cell membrane technologies, including RBC membranes, platelet membranes, macrophage cell membranes, tumour cell membranes, bacterial membranes, hybrid membranes, and their potential for anticancer applications under phototherapy. Lastly, the review discusses the challenges and future directions in this field.
Collapse
Affiliation(s)
- Songtao Li
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xiangrui Meng
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Bo Peng
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Ju Huang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jingwen Liu
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Hang Xiao
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, PR China
| | - Li Ma
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, PR China
| | - Yiyao Liu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, PR China.
| | - Jianyuan Tang
- Traditional Chinese Medicine (TCM) Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
40
|
Meng Y, Chen S, Wang C, Ni X. Advances in Composite Biofilm Biomimetic Nanodrug Delivery Systems for Cancer Treatment. Technol Cancer Res Treat 2024; 23:15330338241250244. [PMID: 38693842 PMCID: PMC11067686 DOI: 10.1177/15330338241250244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/27/2024] [Accepted: 04/08/2024] [Indexed: 05/03/2024] Open
Abstract
Single biofilm biomimetic nanodrug delivery systems based on single cell membranes, such as erythrocytes and cancer cells, have immune evasion ability, good biocompatibility, prolonged blood circulation, and high tumor targeting. Because of the different characteristics and functions of each single cell membrane, more researchers are using various hybrid cell membranes according to their specific needs. This review focuses on several different types of biomimetic nanodrug-delivery systems based on composite biofilms and looks forward to the challenges and possible development directions of biomimetic nanodrug-delivery systems based on composite biofilms to provide reference and ideas for future research.
Collapse
Affiliation(s)
- Yanyan Meng
- School of Pharmacy, Changzhou University, Changzhou, China
- Department of Radiotherapy Oncology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Changzhou Key Laboratory of Medical Physics, Changzhou, China
| | - Shaoqing Chen
- Department of Radiotherapy Oncology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Changzhou Key Laboratory of Medical Physics, Changzhou, China
| | - Cheli Wang
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Xinye Ni
- Department of Radiotherapy Oncology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, China
- Changzhou Key Laboratory of Medical Physics, Changzhou, China
| |
Collapse
|
41
|
Azizollahi F, Kamali H, Oroojalian F. Magnetic nanocarriers for cancer immunotherapy. NANOMEDICINE IN CANCER IMMUNOTHERAPY 2024:349-401. [DOI: 10.1016/b978-0-443-18770-4.00016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
42
|
Chen T, Wang Y, Zhu L, Wu J, Lin J, Huang W, Yan D. Hybrid Membrane Camouflaged Chemodrug-Gene Nanoparticles for Enhanced Combination Therapy of Ovarian Cancer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58067-58078. [PMID: 38056905 DOI: 10.1021/acsami.3c10586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Recently, cell membrane camouflaged nanoparticles (NPs) endowed with natural cellular functions have been extensively studied in various biomedical fields. However, there are few reports about such biomimetic NPs used to codeliver chemodrug and genes for synergistic cancer treatment up to now. Herein, we first prepare chemodrug-gene nanoparticles (Mito-Her2 NPs) by the electrostatic interaction coself-assembly of mitoxantrone hydrochloride (Mito) and human epidermal growth factor receptor-2 antisense oligonucleotide (Her2 ASO). Then, Mito-Her2 NPs are coated by a hybrid membrane (RSHM), consisting of the red blood cell membrane (RBCM) and the SKOV3 ovarian cancer cell membrane (SCM), to produce biomimetic chemodrug-gene nanoparticles (Mito-Her2@RSHM NPs) for combination therapy of ovarian cancer. Mito-Her2@RSHM NPs integrate the advantages of RBCM (e.g., good immune evasion capability and long circulation lifetime in the blood) and SCM (e.g., highly specific cognate recognition) together and improve the anticancer efficacy of Mito-Her2 NPs. The results show that Mito-Her2@RSHM NPs can be devoured by SKOV3 ovarian cancer cells and effectively degraded to release Her2 ASOs and Mito simultaneously. Her2 ASOs can inhibit the expression of endogenous Her2 genes and recover cancer cells' sensitivity to Mito, which ultimately led to a high apoptosis rate of 75.7% in vitro. Mito-Her2@RSHM NPs also show a high tumor suppression rate of 83.33 ± 4.16% in vivo without significant damage to normal tissues. In summary, Mito-Her2@RSHM NPs would be expected as a versatile and safe nanodrug delivery platform with high efficiency for chemo-gene combined cancer treatment.
Collapse
Affiliation(s)
- Tianbao Chen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yuling Wang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Lijuan Zhu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200217, China
| | - Jingchun Wu
- Zhejiang Haobang Chemical Co., LTD, 26 Luyin Road, Quzhou Hi-Tech Industrial Park, Zhejiang 324100, China
| | - Jintang Lin
- Zhejiang Haobang Chemical Co., LTD, 26 Luyin Road, Quzhou Hi-Tech Industrial Park, Zhejiang 324100, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
43
|
Raza F, Zafar H, Jiang L, Su J, Yuan W, Qiu M, Paiva-Santos AC. Progress of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. Biomater Sci 2023; 12:57-91. [PMID: 37902579 DOI: 10.1039/d3bm01170d] [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: 10/31/2023]
Abstract
In recent years, considerable attention has been given to phototherapy, including photothermal and photodynamic therapy to kill tumor cells by producing heat or reactive oxygen species (ROS). It has the high merits of noninvasiveness and limited drug resistance. To fully utilize this therapy, an extraordinary nanovehicle is required to target phototherapeutic agents in the tumor cells. Nanovesicles embody an ideal strategy for drug delivery applications. Cell membrane-derived biomimetic nanovesicles represent a developing type of nanocarrier. Combining this technique with cancer phototherapy could enable a novel strategy. Herein, efforts are made to describe a comprehensive overview of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. The description in this review is mainly based on representative examples of exosome-derived biomimetic nanomedicine research, ranging from their comparison with traditional nanocarriers to extensive applications in cancer phototherapy. Additionally, the challenges and future prospectives for translating these for clinical application are discussed.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Liangdi Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
44
|
Sun M, Yang J, Fan Y, Zhang Y, Sun J, Hu M, Sun K, Zhang J. Beyond Extracellular Vesicles: Hybrid Membrane Nanovesicles as Emerging Advanced Tools for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303617. [PMID: 37749882 PMCID: PMC10646251 DOI: 10.1002/advs.202303617] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/02/2023] [Indexed: 09/27/2023]
Abstract
Extracellular vesicles (EVs), involved in essential physiological and pathological processes of the organism, have emerged as powerful tools for disease treatment owing to their unique natural biological characteristics and artificially acquired advantages. However, the limited targeting ability, insufficient production yield, and low drug-loading capability of natural simplex EVs have greatly hindered their development in clinical translation. Therefore, the establishment of multifunctional hybrid membrane nanovesicles (HMNVs) with favorable adaptability and flexibility has become the key to expanding the practical application of EVs. This timely review summarizes the current progress of HMNVs for biomedical applications. Different HMNVs preparation strategies including physical, chemical, and chimera approaches are first discussed. This review then individually describes the diverse types of HMNVs based on homologous or heterologous cell membrane substances, a fusion of cell membrane and liposome, as well as a fusion of cell membrane and bacterial membrane. Subsequently, a specific emphasis is placed on the highlight of biological applications of the HMNVs toward various diseases with representative examples. Finally, ongoing challenges and prospects of the currently developed HMNVs in clinical translational applications are briefly presented. This review will not only stimulate broad interest among researchers from diverse disciplines but also provide valuable insights for the development of promising nanoplatforms in precision medicine.
Collapse
Affiliation(s)
- Meng Sun
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| | - Jiani Yang
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| | - Yueyun Fan
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| | - Yinfeng Zhang
- International Medical CenterBeijing Friendship HospitalCapital Medical UniversityBeijing100050P. R. China
| | - Jian Sun
- Department of Hepatobiliary SurgeryJinan University First Affiliated HospitalGuangzhou510630P. R. China
| | - Min Hu
- Department of Hepatobiliary SurgeryJinan University First Affiliated HospitalGuangzhou510630P. R. China
| | - Ke Sun
- Department of Urinary surgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| |
Collapse
|
45
|
Li J, Zhang J, Gao Y, Lei S, Wu J, Chen X, Wang K, Duan X, Men K. Targeted siRNA Delivery by Bioinspired Cancer Cell Membrane-Coated Nanoparticles with Enhanced Anti-Cancer Immunity. Int J Nanomedicine 2023; 18:5961-5982. [PMID: 37901359 PMCID: PMC10612485 DOI: 10.2147/ijn.s429036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/07/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Cell-membrane nanocarriers are usually constructed by modifying the nanoparticle surface with cell membrane extracts, which has a direct benefit in endowing targeting capacity to nanocarriers based on their original cell types. However, delivering nucleic acid cargos by cell membrane-based nanoparticles is difficult owing to the strong negative charge of the cell membrane fraction. In this study, we developed a cancer cell membrane-based drug delivery system, the cMDS, for efficient siRNA delivery. Meanwhile, the cancer-specific immune response stimulated by the gene vector itself could offer synergistic anti-cancer ability. Methods The cMDS was prepared by ultrasound, and its transfection efficiency and anti-cancer ability were examined using cultures of CT26 cells. MTT and red blood cell hemolysis tests were performed to assess the safety of cMDS, while its targeted gene delivery and strong immune stimulation were investigated in a subcutaneous tumor model. Moreover, the detailed anti-cancer immune stimulation mechanisms of cMDS are uncovered by protein chip analysis. Results The cMDS was spherical core-shell structure. It showed high transfection efficiency and anti-cancer ability in vitro. In animal experiments, intravenously administered cMDS/siStat3 complex efficiently suppress the growth of colon cancer. Moreover, the result of protein chip analysis suggested that cMDS affect the migration and chemotaxis of immune cells. Conclusion The cMDS shows obvious tumor tissue-specific accumulation properties and strong immune stimulation ability. It is an advanced targeted gene delivery system with potent immunotherapeutic properties.
Collapse
Affiliation(s)
- Jingmei Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Yan Gao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Jieping Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xiaohua Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Kaiyu Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
46
|
Gallo J, Villasante A. Recent Advances in Biomimetic Nanocarrier-Based Photothermal Therapy for Cancer Treatment. Int J Mol Sci 2023; 24:15484. [PMID: 37895165 PMCID: PMC10607206 DOI: 10.3390/ijms242015484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Nanomedicine presents innovative solutions for cancer treatment, including photothermal therapy (PTT). PTT centers on the design of photoactivatable nanoparticles capable of absorbing non-toxic near-infrared light, generating heat within target cells to induce cell death. The successful transition from benchside to bedside application of PTT critically depends on the core properties of nanoparticles responsible for converting light into heat and the surface properties for precise cell-specific targeting. Precisely targeting the intended cells remains a primary challenge in PTT. In recent years, a groundbreaking approach has emerged to address this challenge by functionalizing nanocarriers and enhancing cell targeting. This strategy involves the creation of biomimetic nanoparticles that combine desired biocompatibility properties with the immune evasion mechanisms of natural materials. This review comprehensively outlines various strategies for designing biomimetic photoactivatable nanocarriers for PTT, with a primary focus on its application in cancer therapy. Additionally, we shed light on the hurdles involved in translating PTT from research to clinical practice, along with an overview of current clinical applications.
Collapse
Affiliation(s)
- Juan Gallo
- Advanced Magnetic Theranostic Nanostructures Lab, International Iberian Nanotechnology Laboratory (INL), 4715-330 Braga, Portugal;
| | - Aranzazu Villasante
- Nanobioengineering Lab, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Department of Electronic and Biomedical Engineering, Faculty of Physics, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
47
|
Zong R, Ruan H, Liu C, Fan S, Li J. Bacteria and Bacterial Components as Natural Bio-Nanocarriers for Drug and Gene Delivery Systems in Cancer Therapy. Pharmaceutics 2023; 15:2490. [PMID: 37896250 PMCID: PMC10610331 DOI: 10.3390/pharmaceutics15102490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteria and bacterial components possess multifunctional properties, making them attractive natural bio-nanocarriers for cancer diagnosis and targeted treatment. The inherent tropic and motile nature of bacteria allows them to grow and colonize in hypoxic tumor microenvironments more readily than conventional therapeutic agents and other nanomedicines. However, concerns over biosafety, limited antitumor efficiency, and unclear tumor-targeting mechanisms have restricted the clinical translation and application of natural bio-nanocarriers based on bacteria and bacterial components. Fortunately, bacterial therapies combined with engineering strategies and nanotechnology may be able to reverse a number of challenges for bacterial/bacterial component-based cancer biotherapies. Meanwhile, the combined strategies tend to enhance the versatility of bionanoplasmic nanoplatforms to improve biosafety and inhibit tumorigenesis and metastasis. This review summarizes the advantages and challenges of bacteria and bacterial components in cancer therapy, outlines combinatorial strategies for nanocarriers and bacterial/bacterial components, and discusses their clinical applications.
Collapse
Affiliation(s)
| | | | | | - Shaohua Fan
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| | - Jun Li
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| |
Collapse
|
48
|
Jiang Y, Zhou Z, Liu C, Wang L, Li C. Bacterial outer membrane vesicles as drug delivery carrier for photodynamic anticancer therapy. Front Chem 2023; 11:1284292. [PMID: 37915541 PMCID: PMC10616255 DOI: 10.3389/fchem.2023.1284292] [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: 08/28/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
Photodynamic Therapy (PDT) is an effective tumor treatment strategy that not only induces photocytotoxicity to kill tumor cells directly but also activates the immune system in the body to generate tumor-specific immunity, preventing cancer metastasis and recurrence. However, some limitations of PDT limit the therapeutic efficacy in deep tumors. Previous studies have used different types of nanoparticles (NPs) as drug carriers of photosensitizers (PSs) to overcome the shortcomings of PDT and improve therapeutic efficacy. Among them, bacterial outer membrane vesicles (OMVs) have natural advantages as carriers for PS delivery. In addition to the targeted delivery of PSs into tumor cells, their unique immunogenicity helps them to serve as immune adjuvants to enhance the PDT-induced immune effect, providing new ideas for photodynamic anticancer therapy. Therefore, in this review, we will introduce the biogenesis and anticancer functions of OMVs and the research on them as drug delivery carriers in PDT. Finally, we also discuss the challenges and prospects of OMVs as a versatile drug delivery carrier for photodynamic anticancer therapy.
Collapse
Affiliation(s)
- Yuan Jiang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - ZunZhen Zhou
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Chongzhi Liu
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Limei Wang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Chun Li
- Department of Rehabilitation Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
49
|
Li J, Zhou H, Liu C, Zhang S, Du R, Deng Y, Zou X. Biomembrane‐inspired design of medical micro/nanorobots: From cytomembrane stealth cloaks to cellularized Trojan horses. AGGREGATE 2023; 4. [DOI: 10.1002/agt2.359] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
AbstractMicro/nanorobots are promising for a wide range of biomedical applications (such as targeted tumor, thrombus, and infection therapies in hard‐to‐reach body sites) because of their tiny size and high maneuverability through the actuation of external fields (e.g., magnetic field, light, ultrasound, electric field, and/or heat). However, fully synthetic micro/nanorobots as foreign objects are susceptible to phagocytosis and clearance by diverse phagocytes. To address this issue, researchers have attempted to develop various cytomembrane‐camouflaged micro/nanorobots by two means: (1) direct coating of micro/nanorobots with cytomembranes derived from living cells and (2) the swallowing of micro/nanorobots by living immunocytes via phagocytosis. The camouflaging with cytomembranes or living immunocytes not only protects micro/nanorobots from phagocytosis, but also endows them with new characteristics or functionalities, such as prolonging propulsion in biofluids, targeting diseased areas, or neutralizing bacterial toxins. In this review, we comprehensively summarize the recent advances and developments of cytomembrane‐camouflaged medical micro/nanorobots. We first discuss how cytomembrane coating nanotechnology has been employed to engineer synthetic nanomaterials, and then we review in detail how cytomembrane camouflage tactic can be exploited to functionalize micro/nanorobots. We aim to bridge the gap between cytomembrane‐cloaked micro/nanorobots and nanomaterials and to provide design guidance for developing cytomembrane‐camouflaged micro/nanorobots.
Collapse
Affiliation(s)
- Jinhua Li
- School of Medical Technology Beijing Institute of Technology Beijing China
| | - Huaijuan Zhou
- Advanced Research Institute of Multidisciplinary Sciences Beijing Institute of Technology Beijing China
| | - Chun Liu
- Center for Translational Medicine Precision Medicine Institute The First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology Department of Spinal Surgery The First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Shuailong Zhang
- School of Mechatronical Engineering Beijing Institute of Technology Beijing China
| | - Ran Du
- School of Materials Science & Engineering Key Laboratory of High Energy Density Materials of the Ministry of Education Beijing Institute of Technology Beijing China
| | - Yulin Deng
- School of Life Science Beijing Institute of Technology Beijing China
| | - Xuenong Zou
- Center for Translational Medicine Precision Medicine Institute The First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology Department of Spinal Surgery The First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| |
Collapse
|
50
|
Liu Q, Hu Y, Zheng P, Yang Y, Fu Y, Yang Y, Duan B, Wang M, Li D, Li W, He J, Zheng X, Long Q, Ma Y. Exploiting immunostimulatory mechanisms of immunogenic cell death to develop membrane-encapsulated nanoparticles as a potent tumor vaccine. J Nanobiotechnology 2023; 21:326. [PMID: 37684628 PMCID: PMC10492316 DOI: 10.1186/s12951-023-02031-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/28/2023] [Indexed: 09/10/2023] Open
Abstract
Vaccine is one of the most promising strategies for cancer immunotherapy; however, there are no therapeutic cancer vaccine achieving significant clinical efficacy till now. The main limiting factors include the immune suppression and escape mechanisms developed by tumor and not enough capacity of vaccines to induce a vigorous anti-tumor immunity. This study aimed to develop a strategy of membrane-based biomimetic nanovaccine and investigate the immunological outcomes of utilizing the unique immunostimulatory mechanisms derived of immunogenic cell death (ICD) and of fulfilling a simultaneous nanoscale delivery of a highlighted tumor antigen and broad membrane-associated tumor antigens in the vaccine design. TC-1 tumor cells were treated in vitro with a mixture of mitoxantrone and curcumin for ICD induction, and then chitosan (CS)-coated polylactic co-glycolic acid (PLGA) nanoparticles loaded with HPV16 E744-62 peptides were decorated with the prepared ICD tumor cell membrane (IM); further, the IM-decorated nanoparticles along with adenosine triphosphate (ATP) were embedded with sodium alginate (ALG) hydrogel, And then, the immunological features and therapeutic potency were evaluated in vitro and in vivo. The nanovaccine significantly stimulated the migration, antigen uptake, and maturation of DCs in vitro, improved antigen lysosome escape, and promoted the retention at injection site and accumulation in LNs of the tumor antigen in vivo. In a subcutaneously grafted TC-1 tumor model, the therapeutic immunization of nanovaccine elicited a dramatical antitumor immunity. This study provides a strategy for the development of tumor vaccines.
Collapse
Affiliation(s)
- Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China.
- School of Life Sciences, Yunnan University, Kunming, 650091, China.
| |
Collapse
|