1
|
Li Z, Tan G, Xie H, Lu S. The Application of Regenerated Silk Fibroin in Tissue Repair. MATERIALS (BASEL, SWITZERLAND) 2024; 17:3924. [PMID: 39203101 PMCID: PMC11355482 DOI: 10.3390/ma17163924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024]
Abstract
Silk fibroin (SF) extracted from silk is non-toxic and has excellent biocompatibility and biodegradability, making it an excellent biomedical material. SF-based soft materials, including porous scaffolds and hydrogels, play an important role in accurately delivering drugs to wounds, creating microenvironments for the adhesion and proliferation of support cells, and in tissue remodeling, repair, and wound healing. This article focuses on the study of SF protein-based soft materials, summarizing their preparation methods and basic applications, as well as their regenerative effects, such as drug delivery carriers in various aspects of tissue engineering such as bone, blood vessels, nerves, and skin in recent years, as well as their promoting effects on wound healing and repair processes. The authors expect SF soft materials to play an important role in the field of tissue repair.
Collapse
Affiliation(s)
| | | | | | - Shenzhou Lu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China; (Z.L.); (G.T.); (H.X.)
| |
Collapse
|
2
|
Chen M, Zhang J, Li H, Deng Y, Huang Y, Shen W, Zeng Y, Ci T. Engineered platelet-based immune engager for tumor post-surgery treatment. BIOMATERIALS ADVANCES 2024; 158:213796. [PMID: 38342024 DOI: 10.1016/j.bioadv.2024.213796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/13/2024]
Abstract
Tumor metastasis and recurrence are principal reasons for the high mortality and poor prognosis of cancers. Inefficient engagement between T cell and tumor cell, as well as the universal existence of immune checkpoints, are important factors to the limited immunological surveillance of the immune systems to tumor cells. Herein, an immune engager based on engineered platelets with CD3 antibody modification (P-aCD3) was constructed to facilitate the contact between T cell and tumor cell via providing the anchoring sites of above two cells. Combined with the immune checkpoint blockade strategy, P-aCD3 effectively enhanced T cell mediated cytotoxicity and inhibited tumor recurrence and metastasis in mice melanoma postoperative model and breast cancer model, resulting in significantly prolonged survival of mice.
Collapse
Affiliation(s)
- Mo Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Jinniu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Yueyang Deng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Yun Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenhao Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yixing Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianyuan Ci
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
3
|
Li D, Taylor A, Shi H, Zhou F, Li P, Joshi J, Zhu W, Wang S. Peptide-Guided Nanoparticle Drug Delivery for Cardiomyocytes. BIOLOGY 2024; 13:47. [PMID: 38248477 PMCID: PMC10812947 DOI: 10.3390/biology13010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND Nanoparticles (NPs) have been extensively utilized as a drug delivery system to control the release of therapeutic agents to treat cardiac injuries. However, despite the advantages of utilizing NP-based drug delivery for treating heart diseases, the current delivery system lacks specificity in targeting the cardiac tissue, thus limiting its application. METHODS We created three linear peptides, each consisting of 16-24 amino acids. These peptides were conjugated on the surface of NPs, resulting in the formation of cardiac targeting peptide (CTP)-NPs (designated as CTP-NP1, CTP-NP2, and CTP-NP3). To assess their effectiveness, we compared the binding efficiency of these three CTP-NPs to human and mouse cardiomyocytes. Additionally, we determined their distribution 24 h after injecting the CTP-NPs intravenously into adult C57BL/6J mice. RESULTS When compared to control NPs without CTP (Con-NPs), all three CTP-NPs exhibited significantly increased binding affinity to both human and mouse cardiomyocytes in vitro and enhanced retention in mouse hearts in vivo. A thorough assessment of the heart sections demonstrated that the binding specificity of CTP-NP3 to cardiomyocytes in vivo was significantly greater than that of Con-NPs. None of the three CTP-NPs were proven to cause cardiomyocyte apoptosis. CONCLUSIONS Biocompatible and safe CTP-NP3 can target the heart via binding to cardiomyocytes. This approach of targeting specific molecules-coated NPs may help in delivering therapeutic compounds to cardiomyocytes for the treatment of heart diseases with high efficacy and low toxicity to other tissues.
Collapse
Affiliation(s)
- Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Austin Taylor
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Haiwang Shi
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Fang Zhou
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Jyotsna Joshi
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| |
Collapse
|
4
|
Li DY, Li YM, Lv DY, Deng T, Zeng X, You L, Pang QY, Li Y, Zhu BM. Enhanced interaction between genome-edited mesenchymal stem cells and platelets improves wound healing in mice. J Tissue Eng 2024; 15:20417314241268917. [PMID: 39329066 PMCID: PMC11425747 DOI: 10.1177/20417314241268917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/26/2024] [Indexed: 09/28/2024] Open
Abstract
Impaired wound healing poses a significant burden on the healthcare system and patients. Stem cell therapy has demonstrated promising potential in the treatment of wounds. However, its clinical application is hindered by the low efficiency of cell homing. In this study, we successfully integrated P-selectin glycoprotein ligand-1 (PSGL-1) into the genome of human adipose-derived mesenchymal stem cells (ADSCs) using a Cas9-AAV6-based genome editing tool platform. Our findings revealed that PSGL-1 knock-in enhanced the binding of ADSCs to platelets and their adhesion to the injured site. Moreover, the intravenous infusion of PSGL-1 -engineered ADSCs (KI-ADSCs) significantly improved the homing efficiency and residence rate at the site of skin lesions in mice. Mechanistically, PSGL-1 knock-in promotes the release of some therapeutic cytokines by activating the canonical WNT/β-catenin signaling pathway and accelerates the healing of wounds by promoting angiogenesis, re-epithelialization, and granulation tissue formation at the wound site. This study provides a novel strategy to simultaneously address the problem of poor migration and adhesion of mesenchymal stem cells (MSCs).
Collapse
Affiliation(s)
- De-Yong Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Meng Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan-Yi Lv
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Zeng
- Key Laboratory of Transplant Engineering and Immunology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiu-Yu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Li M, Guo Q, Zhong C, Zhang Z. Multifunctional cell membranes-based nano-carriers for targeted therapies: a review of recent trends and future perspective. Drug Deliv 2023; 30:2288797. [PMID: 38069500 PMCID: PMC10987056 DOI: 10.1080/10717544.2023.2288797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/05/2023] [Indexed: 12/18/2023] Open
Abstract
Nanotechnology has ignited a transformative revolution in disease detection, prevention, management, and treatment. Central to this paradigm shift is the innovative realm of cell membrane-based nanocarriers, a burgeoning class of biomimetic nanoparticles (NPs) that redefine the boundaries of biomedical applications. These remarkable nanocarriers, designed through a top-down approach, harness the intrinsic properties of cell-derived materials as their fundamental building blocks. Through shrouding themselves in natural cell membranes, these nanocarriers extend their circulation longevity and empower themselves to intricately navigate and modulate the multifaceted microenvironments associated with various diseases. This comprehensive review provides a panoramic view of recent breakthroughs in biomimetic nanomaterials, emphasizing their diverse applications in cancer treatment, cardiovascular therapy, viral infections, COVID-19 management, and autoimmune diseases. In this exposition, we deliver a concise yet illuminating overview of the distinctive properties underpinning biomimetic nanomaterials, elucidating their pivotal role in biomedical innovation. We subsequently delve into the exceptional advantages these nanomaterials offer, shedding light on the unique attributes that position them at the forefront of cutting-edge research. Moreover, we briefly explore the intricate synthesis processes employed in creating these biomimetic nanocarriers, shedding light on the methodologies that drive their development.
Collapse
Affiliation(s)
- Mo Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Qiushi Guo
- Pharmacy Department, First Hospital of Jilin University—the Eastern Division, Changchun, China
| | - Chongli Zhong
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Ziyan Zhang
- Department of Orthopedics, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Xi L, Wang L, Zhang M, He C, Yang X, Pang Y, Chen H, Cheng F. TNF-R1 Cellular Nanovesicles Loaded on the Thermosensitive F-127 Hydrogel Enhance the Repair of Scalded Skin. ACS Biomater Sci Eng 2023; 9:5843-5854. [PMID: 37043416 PMCID: PMC10566511 DOI: 10.1021/acsbiomaterials.2c01257] [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: 10/24/2022] [Accepted: 02/13/2023] [Indexed: 04/13/2023]
Abstract
Excessive inflammatory response after severe scalding is an important cause of delayed wound healing and is even life-threatening. Tumor necrosis factor α (TNF-α) is a key pro-inflammatory factor of skin trauma. Interacting with tumor necrosis factor receptor 1 (TNF-R1), TNF-α causes excessive inflammation and poor prognosis by activating NF-κB pathway. Antagonizing high levels of TNF-α is one of the therapeutic approaches for diseases associated with the overactivation of inflammatory responses. However, the available monoclonal antibodies are limited in their application due to their complex preparation process, high price, and the lack of cell targeting ability leading to systemic toxicity and side effects. In this study, by using a genetic bioengineering technique, we modified TNF-R1 on the cell membrane surface-derived nanovesicles (NVs). We confirmed that TNF-R1 NVs stably expressed TNF-R1 on the membrane surface and interacted with its ligand TNF-α. Furthermore, TNF-R1 NVs competitively antagonized the effect of TNF-α in the wound healing assay in vitro. In the scalded mouse model, TNF-R1 NVs were released continuously from the thermosensitive hydrogel Pluronic F-127, resulting in less inflammation and better wound healing. Our results revealed TNF-R1 NVs as promising cell-free therapeutic agents in alleviating TNF-α-mediated pro-inflammatory signaling and promoting wound repair.
Collapse
Affiliation(s)
- Lifang Xi
- School
of Pharmaceutical Sciences (Shenzhen), Shenzhen
Campus of Sun Yat-sen University, Shenzhen, China, 518107
| | - Linglu Wang
- School
of Pharmaceutical Sciences (Shenzhen), Shenzhen
Campus of Sun Yat-sen University, Shenzhen, China, 518107
| | - Manqi Zhang
- School
of Pharmaceutical Sciences (Shenzhen), Shenzhen
Campus of Sun Yat-sen University, Shenzhen, China, 518107
| | - Chao He
- School
of Pharmaceutical Sciences (Shenzhen), Shenzhen
Campus of Sun Yat-sen University, Shenzhen, China, 518107
| | - Xinrui Yang
- School
of Pharmaceutical Sciences (Shenzhen), Shenzhen
Campus of Sun Yat-sen University, Shenzhen, China, 518107
| | - Yuxin Pang
- School
of Pharmacy, Guizhou University of Traditional
Chinese Medicine, Guiyang, Guizhou, 550025, China
| | - Hongbo Chen
- School
of Pharmaceutical Sciences (Shenzhen), Shenzhen
Campus of Sun Yat-sen University, Shenzhen, China, 518107
| | - Fang Cheng
- School
of Pharmaceutical Sciences (Shenzhen), Shenzhen
Campus of Sun Yat-sen University, Shenzhen, China, 518107
| |
Collapse
|
7
|
Liu WS, Wu LL, Chen CM, Zheng H, Gao J, Lu ZM, Li M. Lipid-hybrid cell-derived biomimetic functional materials: A state-of-the-art multifunctional weapon against tumors. Mater Today Bio 2023; 22:100751. [PMID: 37636983 PMCID: PMC10448342 DOI: 10.1016/j.mtbio.2023.100751] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Tumors are among the leading causes of death worldwide. Cell-derived biomimetic functional materials have shown great promise in the treatment of tumors. These materials are derived from cell membranes, extracellular vesicles and bacterial outer membrane vesicles and may evade immune recognition, improve drug targeting and activate antitumor immunity. However, their use is limited owing to their low drug-loading capacity and complex preparation methods. Liposomes are artificial bionic membranes that have high drug-loading capacity and can be prepared and modified easily. Although they can overcome the disadvantages of cell-derived biomimetic functional materials, they lack natural active targeting ability. Lipids can be hybridized with cell membranes, extracellular vesicles or bacterial outer membrane vesicles to form lipid-hybrid cell-derived biomimetic functional materials. These materials negate the disadvantages of both liposomes and cell-derived components and represent a promising delivery platform in the treatment of tumors. This review focuses on the design strategies, applications and mechanisms of action of lipid-hybrid cell-derived biomimetic functional materials and summarizes the prospects of their further development and the challenges associated with it.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| | - Li-Li Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Cui-Min Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Hao Zheng
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| |
Collapse
|
8
|
Yin T, Liu Y, He B, Gong B, Chu J, Gao C, Liang W, Hao M, Sun W, Zhuang J, Gao J, Yin Y. Cell primitive-based biomimetic nanomaterials for Alzheimer's disease targeting and therapy. Mater Today Bio 2023; 22:100789. [PMID: 37706205 PMCID: PMC10495673 DOI: 10.1016/j.mtbio.2023.100789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/15/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, which is not just confined to the older population. Although developments have been made in AD treatment, various limitations remain to be addressed. These are partly contributed by biological hurdles, such as the blood-brain barrier and peripheral side effects, as well as by lack of carriers that can efficiently deliver the therapeutics to the brain while preserving their therapeutic efficacy. The increasing AD prevalence and the unavailability of effective treatments have encouraged researchers to develop improved, convenient, and affordable therapies. Functional materials based on primitive cells and nanotechnology are emerging as attractive therapeutics in AD treatment. Cell primitives possess distinct biological functions, including long-term circulation, lesion site targeting, and immune suppression. This review summarizes the challenges in the delivery of AD drugs and recent advances in cell primitive-based materials for AD treatment. Various cell primitives, such as cells, extracellular vesicles, and cell membranes, are presented together with their distinctive biological functions and construction strategies. Moreover, future research directions are discussed on the basis of foreseeable challenges and perspectives.
Collapse
Affiliation(s)
- Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Clinical pharmacy innovation institute, Shanghai Jiao Tong University of Medicine, Shanghai, 200000, China
| | - Bin He
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Baofeng Gong
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Chao Gao
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
- School of Health Science and Engineering, University of Shanghaifor Science and Technology, Shanghai, 200093, China
| | - Mengqi Hao
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
- School of Health Science and Engineering, University of Shanghaifor Science and Technology, Shanghai, 200093, China
| | - Wenjing Sun
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, 200003, China
| |
Collapse
|
9
|
Zeng S, Tang Q, Xiao M, Tong X, Yang T, Yin D, Lei L, Li S. Cell membrane-coated nanomaterials for cancer therapy. Mater Today Bio 2023; 20:100633. [PMID: 37128288 PMCID: PMC10148189 DOI: 10.1016/j.mtbio.2023.100633] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/01/2023] [Accepted: 04/09/2023] [Indexed: 05/03/2023] Open
Abstract
With the development of nanotechnology, nanoparticles have emerged as a delivery carrier for tumor drug therapy, which can improve the therapeutic effect by increasing the stability and solubility and prolonging the half-life of drugs. However, nanoparticles are foreign substances for humans, are easily cleared by the immune system, are less targeted to tumors, and may even be toxic to the body. As a natural biological material, cell membranes have unique biological properties, such as good biocompatibility, strong targeting ability, the ability to evade immune surveillance, and high drug-carrying capacity. In this article, we review cell membrane-coated nanoparticles (CMNPs) and their applications to tumor therapy. First, we briefly describe CMNP characteristics and applications. Second, we present the characteristics and advantages of different cell membranes as well as nanoparticles, provide a brief description of the process of CMNPs, discuss the current status of their application to tumor therapy, summarize their shortcomings for use in cancer therapy, and propose future research directions. This review summarizes the research progress on CMNPs in cancer therapy in recent years and assesses remaining problems, providing scholars with new ideas for future research on CMNPs in tumor therapy.
Collapse
Affiliation(s)
- Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xinying Tong
- Department of Hemodialysis, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Tao Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Danhui Yin
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Corresponding author.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Corresponding author.
| |
Collapse
|
10
|
Chen BQ, Zhao Y, Zhang Y, Pan YJ, Xia HY, Kankala RK, Wang SB, Liu G, Chen AZ. Immune-regulating camouflaged nanoplatforms: A promising strategy to improve cancer nano-immunotherapy. Bioact Mater 2023; 21:1-19. [PMID: 36017071 PMCID: PMC9382433 DOI: 10.1016/j.bioactmat.2022.07.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/11/2022] [Accepted: 07/24/2022] [Indexed: 02/06/2023] Open
Abstract
Although nano-immunotherapy has advanced dramatically in recent times, there remain two significant hurdles related to immune systems in cancer treatment, such as (namely) inevitable immune elimination of nanoplatforms and severely immunosuppressive microenvironment with low immunogenicity, hampering the performance of nanomedicines. To address these issues, several immune-regulating camouflaged nanocomposites have emerged as prevailing strategies due to their unique characteristics and specific functionalities. In this review, we emphasize the composition, performances, and mechanisms of various immune-regulating camouflaged nanoplatforms, including polymer-coated, cell membrane-camouflaged, and exosome-based nanoplatforms to evade the immune clearance of nanoplatforms or upregulate the immune function against the tumor. Further, we discuss the applications of these immune-regulating camouflaged nanoplatforms in directly boosting cancer immunotherapy and some immunogenic cell death-inducing immunotherapeutic modalities, such as chemotherapy, photothermal therapy, and reactive oxygen species-mediated immunotherapies, highlighting the current progress and recent advancements. Finally, we conclude the article with interesting perspectives, suggesting future tendencies of these innovative camouflaged constructs towards their translation pipeline.
Collapse
Affiliation(s)
- Biao-Qi Chen
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Yi Zhao
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, PR China
| | - Yu-Jing Pan
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, PR China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| |
Collapse
|
11
|
Ibrahim A, Khalil IA, Mahmoud MY, Bakr AF, Ghoniem MG, Al-Farraj ES, El-Sherbiny IM. Layer-by-layer development of chitosan/alginate-based platelet-mimicking nanocapsules for augmenting doxorubicin cytotoxicity against breast cancer. Int J Biol Macromol 2023; 225:503-517. [PMID: 36403763 DOI: 10.1016/j.ijbiomac.2022.11.107] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/20/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022]
Abstract
Breast carcinoma is considered one of the most invasive and life-threatening malignancies in females. Mastectomy, radiation therapy, hormone therapy and chemotherapy are the most common treatment choices for breast cancer. Doxorubicin (DOX) is one of the most regularly utilized medications in breast cancer protocols. However, DOX has showed numerous side effects including lethal cardiotoxicity. This study aims to fortify DOX cytotoxicity and lowering its side effects via its combining with the antidiabetic metformin (MET) as an adjuvant therapy, along with its effective delivery using natural platelet-rich plasma (PRP), and newly-developed PRP-mimicking nanocapsules (NCs). The PRP-mimicking NCs were fabricated via layer-by-layer (LBL) deposition of oppositely charged biodegradable and biocompatible chitosan (CS) and alginate (ALG) on a core of synthesized polystyrene nanoparticles (PS NPs) followed by removal of the PS core. Both natural PRP and PRP-mimicking NCs were loaded with DOX and MET adjuvant therapy, followed by their physicochemical characterizations including DLS, FTIR, DSC, and morphological evaluation using TEM. In-vitro drug release studies, cytotoxicity, apoptosis/necrosis, and cell cycle analysis were conducted using MCF-7 breast cancer cells. Also, an in-vivo assessment was carried out using EAC-bearing balb/c mice animal model to evaluate the effect of DOX/MET-loaded natural PRP and PRP-mimicked NCs on tumor weight, volume and growth biomarkers in addition to analyzing the immunohistopathology of the treated tissues. Results confirmed the development of CS/ALG-based PRP-mimicking NCs with a higher loading capacity of both drugs (DOX and MET) and smaller size (259.7 ± 19.3 nm) than natural PRP (489 ± 20.827 nm). Both in-vitro and in-vivo studies were in agreement and confirmed that MET synergized the anticancer activity of DOX against breast cancer. Besides, the developed LBL NCs successfully mimicked the PRP in improving the loaded drugs biological efficiency more than free drugs.
Collapse
Affiliation(s)
- Alaa Ibrahim
- Nanomedicine Research Labs, Center for Materials Sciences, Zewail City of Science and Technology, 6th of October City, 12578 Giza, Egypt
| | - Islam A Khalil
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University of Science and Technology (MUST), 6th of October, Giza 12582, Egypt
| | - Mohamed Y Mahmoud
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Alaa F Bakr
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Monira G Ghoniem
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13623, Saudi Arabia
| | - Eida S Al-Farraj
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13623, Saudi Arabia
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Sciences, Zewail City of Science and Technology, 6th of October City, 12578 Giza, Egypt.
| |
Collapse
|
12
|
Li D, Son Y, Jang M, Wang S, Zhu W. Nanoparticle Based Cardiac Specific Drug Delivery. BIOLOGY 2023; 12:biology12010082. [PMID: 36671774 PMCID: PMC9856055 DOI: 10.3390/biology12010082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Heart failure secondary to myocardial injuries is a leading cause of death worldwide. Recently, a growing number of novel therapies have emerged for injured myocardium repairment. However, delivering therapeutic agents specifically to the injured heart remains a significant challenge. Nanoparticles are the most commonly used vehicles for targeted drug delivery. Various nanoparticles have been synthesized to deliver drugs and other therapeutic molecules to the injured heart via passive or active targeting approaches, and their targeting specificity and therapeutic efficacies have been investigated. Here, we summarized nanoparticle-based, cardiac-specific drug delivery systems, their potency for treating heart diseases, and the mechanisms underlying these cardiac-targeting strategies. We also discussed the clinical studies that have employed nanoparticle-based cardiac-specific drug delivery.
Collapse
Affiliation(s)
- Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Department of Cardiology, Dongfang Hospital, The Second Affiliated Hospital of Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yura Son
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- Correspondence: (S.W.); (W.Z.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Correspondence: (S.W.); (W.Z.)
| |
Collapse
|
13
|
Rodrigues CF, Fernandes N, de Melo‐Diogo D, Correia IJ, Moreira AF. Cell-Derived Vesicles for Nanoparticles' Coating: Biomimetic Approaches for Enhanced Blood Circulation and Cancer Therapy. Adv Healthc Mater 2022; 11:e2201214. [PMID: 36121767 PMCID: PMC11481079 DOI: 10.1002/adhm.202201214] [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: 05/23/2022] [Revised: 09/11/2022] [Indexed: 01/28/2023]
Abstract
Cancer nanomedicines are designed to encapsulate different therapeutic agents, prevent their premature release, and deliver them specifically to cancer cells, due to their ability to preferentially accumulate in tumor tissue. However, after intravenous administration, nanoparticles immediately interact with biological components that facilitate their recognition by the immune system, being rapidly removed from circulation. Reports show that less than 1% of the administered nanoparticles effectively reach the tumor site. This suboptimal pharmacokinetic profile is pointed out as one of the main factors for the nanoparticles' suboptimal therapeutic effectiveness and poor translation to the clinic. Therefore, an extended blood circulation time may be crucial to increase the nanoparticles' chances of being accumulated in the tumor and promote a site-specific delivery of therapeutic agents. For that purpose, the understanding of the forces that govern the nanoparticles' interaction with biological components and the impact of the physicochemical properties on the in vivo fate will allow the development of novel and more effective nanomedicines. Therefore, in this review, the nano-bio interactions are summarized. Moreover, the application of cell-derived vesicles for extending the blood circulation time and tumor accumulation is reviewed, focusing on the advantages and shortcomings of each cell source.
Collapse
Affiliation(s)
- Carolina F. Rodrigues
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Natanael Fernandes
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Duarte de Melo‐Diogo
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - Ilídio J. Correia
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
| | - André F. Moreira
- CICS‐UBI – Health Sciences Research CentreUniversidade da Beira InteriorAv. Infante D. HenriqueCovilhã6200‐506Portugal
- CPIRN‐UDI/IPG – Center of Potential and Innovation in Natural Resources, Research Unit for Inland DevelopmentInstituto Politécnico da GuardaAvenida Dr. Francisco de Sá CarneiroGuarda6300‐559Portugal
| |
Collapse
|
14
|
|
15
|
Thangudu S, Huang EY, Su CH. Safe magnetic resonance imaging on biocompatible nanoformulations. Biomater Sci 2022; 10:5032-5053. [PMID: 35858468 DOI: 10.1039/d2bm00692h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Magnetic resonance imaging (MRI) holds promise for the early clinical diagnosis of various diseases, but most clinical MR techniques require the use of a contrast medium. Several nanomaterial (NM) mediated contrast agents (CAs) are widely used as T1- and T2-based MR contrast agents for clinical and non-clinical applications. Unfortunately, most NM-based CAs are toxic or non-biocompatible, restricting their practical/clinical applications. Therefore, the development of nontoxic and biocompatible CAs for clinical MRI diagnosis is highly desired. To this end, several biocompatible and biomimetic strategies have been developed to offer long blood circulation time, significant biocompatibility, in vivo biodistribution and high contrast ability for efficient imaging. However, detailed review reports on biocompatible NMs, specifically for MR imaging have not yet been summarized. Thus, in the present review we summarize various surface coating strategies (such as polymers, proteins, cell membranes, etc.) to achieve biocompatible NPs, providing a detailed discussion of advances and future prospects for safe MRI imaging.
Collapse
Affiliation(s)
- Suresh Thangudu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
| | - Eng-Yen Huang
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chia-Hao Su
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan. .,Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
16
|
Platelet-Membrane-Encapsulated Carvedilol with Improved Targeting Ability for Relieving Myocardial Ischemia-Reperfusion Injury. MEMBRANES 2022; 12:membranes12060605. [PMID: 35736311 PMCID: PMC9227294 DOI: 10.3390/membranes12060605] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/26/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023]
Abstract
In recent years, cell membrane drug delivery systems have received increasing attention. However, drug-loaded membrane delivery systems targeting therapy in myocardial ischemia–reperfusion injury (MIRI) have been relatively rarely studied. The purpose of this study was to explore the protective effect of platelet-membrane-encapsulated Carvedilol on MIRI. We extracted platelets from the blood of adult SD rats and prepared platelet membrane vesicles (PMVs). Carvedilol, a nonselective β-blocker, was encapsulated into the PMVs. In order to determine the best encapsulation rate and drug-loading rate, three different concentrations of Carvedilol in low, medium, and high amounts were fused to the PMVs in different volume ratios (drugs/PMVs at 2:1, 1:1, 1:2, and 4:1) for determining the optimum concentration and volume ratio. By comparing other delivery methods, including abdominal injection and intravenous administration, the efficacy of PMVs-encapsulated drug-targeted delivery treatment was observed. The PMVs have the ability to target ischemic-damaged myocardial tissue, and the concentration and volume ratio at the optimum encapsulation rate and the drug-loading rate are 0.5 mg and 1:1. We verified that PMVs@Carvedilol had better therapeutic effects compared to other treatment groups, and immunofluorescence observation showed a significant improvement in the apoptosis indicators and infarction area of myocardial cells. Targeted administration of PMVs@Carvedilol may be a promising treatment for myocardial reperfusion injury, as it significantly improves postinjury cardiac function and increases drug utilization compared to other delivery methods.
Collapse
|
17
|
Li F, Liu D, Liu M, Ji Q, Zhang B, Mei Q, Cheng Y, Zhou S. Tregs biomimetic nanoparticle to reprogram inflammatory and redox microenvironment in infarct tissue to treat myocardial ischemia reperfusion injury in mice. J Nanobiotechnology 2022; 20:251. [PMID: 35659239 PMCID: PMC9164893 DOI: 10.1186/s12951-022-01445-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/28/2022] [Indexed: 12/11/2022] Open
Abstract
Background At present, patients with myocardial infarction remain an increased risk for myocardial ischemia/reperfusion injury (MI/RI). There lacks effectively method to treat MI/RI in clinic. For the treatment of MI/RI, it is still a bottleneck to effectively deliver drug to ischemic myocardium. In this paper, a regulatory T cells (Tregs) biomimetic nanoparticle (CsA@PPTK) was prepared by camouflaging nanoparticle with platelet membrane. Results CsA@PPTK actively accumulated in ischemic myocardium of MI/RI mice. CsA@PPTK significantly scavenged reactive oxygen species (ROS) and increased the generation of Tregs and the ratio of M2 type macrophage to M1 type macrophage in ischemic myocardium. Moreover, CsA@PPTK significantly attenuated apoptosis of cardiomyocytes and reduced the infarct size and fibrosis area in ischemic myocardium. CsA@PPTK markedly decreased the protein expression of MMP-9 and increased the protein expression of CX43 in ischemic myocardium tissue. Subsequently, the remodeling of the left ventricle was significant alleviated, and heart function of MI/RI mice was markedly improved. Conclusion CsA@PPTK showed significant therapeutic effect on MI/RI, and it has great potential application in the treatment of MI/RI. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01445-2.
Collapse
Affiliation(s)
- Fangyuan Li
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qibing Mei
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China.
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China. .,Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
18
|
Zoulikha M, Huang F, Wu Z, He W. COVID-19 inflammation and implications in drug delivery. J Control Release 2022; 346:260-274. [PMID: 35469984 PMCID: PMC9045711 DOI: 10.1016/j.jconrel.2022.04.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/15/2022] [Indexed: 01/09/2023]
Abstract
Growing evidence indicates that hyperinflammatory syndrome and cytokine storm observed in COVID-19 severe cases are narrowly associated with the disease's poor prognosis. Therefore, targeting the inflammatory pathways seems to be a rational therapeutic strategy against COVID-19. Many anti-inflammatory agents have been proposed; however, most of them suffer from poor bioavailability, instability, short half-life, and undesirable biodistribution resulting in off-target effects. From a pharmaceutical standpoint, the implication of COVID-19 inflammation can be exploited as a therapeutic target and/or a targeting strategy against the pandemic. First, the drug delivery systems can be harnessed to improve the properties of anti-inflammatory agents and deliver them safely and efficiently to their therapeutic targets. Second, the drug carriers can be tailored to develop smart delivery systems able to respond to the microenvironmental stimuli to release the anti-COVID-19 therapeutics in a selective and specific manner. More interestingly, some biosystems can simultaneously repress the hyperinflammation due to their inherent anti-inflammatory potency and endow their drug cargo with a selective delivery to the injured sites.
Collapse
Affiliation(s)
- Makhloufi Zoulikha
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Feifei Huang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
19
|
Rampado R, Caliceti P, Agostini M. Latest Advances in Biomimetic Cell Membrane-Coated and Membrane-Derived Nanovectors for Biomedical Applications. NANOMATERIALS 2022; 12:nano12091543. [PMID: 35564251 PMCID: PMC9104043 DOI: 10.3390/nano12091543] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023]
Abstract
In the last decades, many nanovectors were developed for different diagnostic or therapeutic purposes. However, most nanosystems have been designed using a “bottom-up” approach, in which the basic components of the nanovector become assembled to achieve complex and specific behaviors. Despite the fine control of formulative conditions, the complexity of these systems often results cumbersome and difficult to scale-up. Recently, biomimetic materials emerged as a complementary or alternative design approach through a “top-down strategy”, using cell-derived materials as building blocks to formulate innovative nanovectors. The use of cell membranes as nanoparticle coatings endows nanomaterials with the biological identity and some of the functions of the cells they are derived from. In this review, we discuss some of the latest examples of membrane coated and membrane-derived biomimetic nanomaterials and underline the common general functions offered by the biomaterials used. From these examples, we suggest a systematic classification of these biomimetic materials based on their biological sources and formulation techniques, with their respective advantages and disadvantages, and summarize the current technologies used for membranes isolation and integration on nanovectors. We also discuss some current technical limitations and hint to future direction of the improvement for biomimetics.
Collapse
Affiliation(s)
- Riccardo Rampado
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo, 5, 35131 Padua, Italy; (R.R.); (P.C.)
- Nano-Inspired Biomedicine Lab, Insitute of Pediatric Research-Città della Speranza, Corso Stati Uniti 4, 35127 Padua, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo, 5, 35131 Padua, Italy; (R.R.); (P.C.)
| | - Marco Agostini
- Nano-Inspired Biomedicine Lab, Insitute of Pediatric Research-Città della Speranza, Corso Stati Uniti 4, 35127 Padua, Italy
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Via Nicolò Giustiniani 2, 35128 Padua, Italy
- Correspondence:
| |
Collapse
|
20
|
Li Z, Ding Y, Liu J, Wang J, Mo F, Wang Y, Chen-Mayfield TJ, Sondel PM, Hong S, Hu Q. Depletion of tumor associated macrophages enhances local and systemic platelet-mediated anti-PD-1 delivery for post-surgery tumor recurrence treatment. Nat Commun 2022; 13:1845. [PMID: 35387972 PMCID: PMC8987059 DOI: 10.1038/s41467-022-29388-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 03/15/2022] [Indexed: 02/06/2023] Open
Abstract
Immunosuppressive cells residing in the tumor microenvironment, especially tumor associated macrophages (TAMs), hinder the infiltration and activation of T cells, limiting the anti-cancer outcomes of immune checkpoint blockade. Here, we report a biocompatible alginate-based hydrogel loaded with Pexidartinib (PLX)-encapsulated nanoparticles that gradually release PLX at the tumor site to block colony-stimulating factor 1 receptors (CSF1R) for depleting TAMs. The controlled TAM depletion creates a favorable milieu for facilitating local and systemic delivery of anti-programmed cell death protein 1 (aPD-1) antibody-conjugated platelets to inhibit post-surgery tumor recurrence. The tumor immunosuppressive microenvironment is also reprogrammed by TAM elimination, further promoting the infiltration of T cells into tumor tissues. Moreover, the inflammatory environment after surgery could trigger the activation of platelets to facilitate the release of aPD-1 accompanied with platelet-derived microparticles binding to PD-1 receptors for re-activating T cells. All these results collectively indicate that the immunotherapeutic efficacy against tumor recurrence of both local and systemic administration of aPD-1 antibody-conjugated platelets could be strengthened by local depletion of TAMs through the hydrogel reservoir. Increased density of tumor associated macrophages has been correlated with tumor recurrence following surgery. Here the authors design an alginate-based hydrogel encapsulating anti-PD-1-conjugated platelets and nanoparticles loaded with the macrophage-depleting CSF-1R inhibitor pexidartinib, showing inhibition of post-surgery tumor recurrence in preclinical models.
Collapse
Affiliation(s)
- Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yingyue Ding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jun Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jianxin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Fanyi Mo
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Ting-Jing Chen-Mayfield
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Paul M Sondel
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA. .,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
21
|
Li R, Liu K, Huang X, Li D, Ding J, Liu B, Chen X. Bioactive Materials Promote Wound Healing through Modulation of Cell Behaviors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105152. [PMID: 35138042 PMCID: PMC8981489 DOI: 10.1002/advs.202105152] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/24/2021] [Indexed: 05/13/2023]
Abstract
Skin wound repair is a multistage process involving multiple cellular and molecular interactions, which modulate the cell behaviors and dynamic remodeling of extracellular matrices to maximize regeneration and repair. Consequently, abnormalities in cell functions or pathways inevitably give rise to side effects, such as dysregulated inflammation, hyperplasia of nonmigratory epithelial cells, and lack of response to growth factors, which impedes angiogenesis and fibrosis. These issues may cause delayed wound healing or even non-healing states. Current clinical therapeutic approaches are predominantly dedicated to preventing infections and alleviating topical symptoms rather than addressing the modulation of wound microenvironments to achieve targeted outcomes. Bioactive materials, relying on their chemical, physical, and biological properties or as carriers of bioactive substances, can affect wound microenvironments and promote wound healing at the molecular level. By addressing the mechanisms of wound healing from the perspective of cell behaviors, this review discusses how bioactive materials modulate the microenvironments and cell behaviors within the wounds during the stages of hemostasis, anti-inflammation, tissue regeneration and deposition, and matrix remodeling. A deeper understanding of cell behaviors during wound healing is bound to promote the development of more targeted and efficient bioactive materials for clinical applications.
Collapse
Affiliation(s)
- Ruotao Li
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Kai Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Xu Huang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Di Li
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Bin Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
22
|
Wang X, Zhang W. The Janus of Protein Corona on nanoparticles for tumor targeting, immunotherapy and diagnosis. J Control Release 2022; 345:832-850. [PMID: 35367478 DOI: 10.1016/j.jconrel.2022.03.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/11/2022]
Abstract
The therapeutics based on nanoparticles (NPs) are considered as the promising strategy for tumor detection and treatment. However, one of the most challenges is the adsorption of biomolecules on NPs after their exposition to biological medium, leading unpredictable in vivo behaviors. The interactions caused by protein corona (PC) will influence the biological fate of NPs in either negative or positive ways, including (i) blood circulation, accumulation and penetration of NPs at targeting sites, and further cellular uptake in tumor targeting delivery; (ii) interactions between NPs and receptors on immune cells for immunotherapy. Besides, PC on NPs could be utilized as new biomarker in tumor diagnosis by identifying the minor change of protein concentration led by tumor growth and invasion in blood. Herein, the mechanisms of these PC-mediated effects will be introduced. Moreover, the recent advances about the strategies will be reviewed to reduce negative effects caused by PC and/or utilize positive effects of PC on tumor targeting, immunotherapy and diagnosis, aiming to provide a reasonable perspective to recognize PC with their applications.
Collapse
Affiliation(s)
- Xiaobo Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wenli Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
23
|
Qiao K, Xu L, Tang J, Wang Q, Lim KS, Hooper G, Woodfield TBF, Liu G, Tian K, Zhang W, Cui X. The advances in nanomedicine for bone and cartilage repair. J Nanobiotechnology 2022; 20:141. [PMID: 35303876 PMCID: PMC8932118 DOI: 10.1186/s12951-022-01342-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/01/2022] [Indexed: 12/24/2022] Open
Abstract
With the gradual demographic shift toward an aging and obese society, an increasing number of patients are suffering from bone and cartilage injuries. However, conventional therapies are hindered by the defects of materials, failing to adequately stimulate the necessary cellular response to promote sufficient cartilage regeneration, bone remodeling and osseointegration. In recent years, the rapid development of nanomedicine has initiated a revolution in orthopedics, especially in tissue engineering and regenerative medicine, due to their capacity to effectively stimulate cellular responses on a nanoscale with enhanced drug loading efficiency, targeted capability, increased mechanical properties and improved uptake rate, resulting in an improved therapeutic effect. Therefore, a comprehensive review of advancements in nanomedicine for bone and cartilage diseases is timely and beneficial. This review firstly summarized the wide range of existing nanotechnology applications in the medical field. The progressive development of nano delivery systems in nanomedicine, including nanoparticles and biomimetic techniques, which are lacking in the current literature, is further described. More importantly, we also highlighted the research advancements of nanomedicine in bone and cartilage repair using the latest preclinical and clinical examples, and further discussed the research directions of nano-therapies in future clinical practice.
Collapse
Affiliation(s)
- Kai Qiao
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Lu Xu
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
- Department of Dermatology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 61004, Sichuan, China
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Gary Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Guozhen Liu
- School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, 518172, Guangdong, China
| | - Kang Tian
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
| | - Weiguo Zhang
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
| | - Xiaolin Cui
- Department of Bone & Joint, the First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand.
| |
Collapse
|
24
|
Zhou J, Niu C, Huang B, Chen S, Yu C, Cao S, Pei W, Guo R. Platelet Membrane Biomimetic Nanoparticles Combined With UTMD to Improve the Stability of Atherosclerotic Plaques. Front Chem 2022; 10:868063. [PMID: 35350774 PMCID: PMC8958035 DOI: 10.3389/fchem.2022.868063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
Although research on the treatment of atherosclerosis has progressed recently, challenges remain in developing more effective, safer and transformative strategies for the treatment of atherosclerosis. Nanomaterials have recently played a unique role in many fields, including atherosclerosis treatment. Platelets are common component in the blood. Due to their inherent properties, platelets can target and adhere to atherosclerotic plaques. Ultrasound-targeted microbubble destruction (UTMD) shows great prospects in promoting the efficiency of drug delivery in treating solid tumors. In this study, we explored the possibility that UTMD assists platelet biomimetic rapamycin (RAP)-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles (RAP@PLT NPs) in the treatment of atherosclerosis. The biomimetic nano-formulations exhibit better targeting ability to plaques when administered in vivo. Targeted destruction of Sonovue™ in the aortic area further improved the efficiency of targeting plaques. Moreover, the progression of atherosclerotic plaques was inhibited, and the stability of plaques was improved. Together, our study established a novel strategy for targeted delivery of nanoparticles in atherosclerotic plaques, by combining the advantages of the ultrasonic cavitation effect and biomimicking nanoparticles in drug delivery.
Collapse
Affiliation(s)
- Jia Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Ultrasound Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Biying Huang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sijie Chen
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Caigui Yu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Sheng Cao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Sheng Cao, ; Wenjing Pei, ; Ruiqiang Guo,
| | - Wenjing Pei
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Sheng Cao, ; Wenjing Pei, ; Ruiqiang Guo,
| | - Ruiqiang Guo
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Sheng Cao, ; Wenjing Pei, ; Ruiqiang Guo,
| |
Collapse
|
25
|
Wang X, Meng X, Meng L, Guo Y, Li Y, Yang C, Pei Z, Li J, Wang F. Joint efficacy of the three biomarkers SNCA, GYPB and HBG1 for atrial fibrillation and stroke: Analysis via the support vector machine neural network. J Cell Mol Med 2022; 26:2010-2022. [PMID: 35138035 PMCID: PMC8980947 DOI: 10.1111/jcmm.17224] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/13/2022] [Accepted: 01/24/2022] [Indexed: 12/29/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of persistent arrhythmia. Although its incidence has been increasing, the pathogenesis of AF in stroke remains unclear. In this study, a total of 30 participants were recruited, including 10 controls, 10 patients with AF and 10 patients with AF and stroke (AF + STROKE). Differentially expressed genes (DEGs) were identified, and functional annotation of DEGs, comparative toxicogenomic database analysis associated with cardiovascular diseases, and predictions of miRNAs of hub genes were performed. Using RT‐qPCR, biological process and support vector machine neural networks, numerous DEGs were found to be related to AF. HBG1, SNCA and GYPB were found to be upregulated in the AF group. Higher expression of hub genes in AF and AF + STROKE groups was detected via RT‐PCR. Upon training the biological process neural network of SNCA and GYPB for HBG1, only small differences were detected. Based on the support vector machine, the predicted value of SNCA and GYPB for HBG1 was 0.9893. Expression of the hub genes of HBG1, SNCA and GYPB might therefore be significantly correlated to AF. These genes are involved in the incidence of AF complicated by stroke, and may serve as targets for early diagnosis and treatment.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuyang Meng
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lingbing Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Guo
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chenguang Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zuowei Pei
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiahan Li
- The First Mobile Corps of People's Armed Police, Beijing, China
| | - Fang Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
26
|
Sun Z, Chen J, Chen G, Zhang C, Li C. Recent advances of engineered and artificial drug delivery system towards solid tumor based on immune cells. Biomed Mater 2022; 17. [PMID: 35042206 DOI: 10.1088/1748-605x/ac4c8b] [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: 09/25/2021] [Accepted: 01/18/2022] [Indexed: 11/11/2022]
Abstract
Precise drug delivery in cancer treatment is a long-standing concern of modern medicine. Compared with traditional molecular medicines and nano-medicines, emerging cell-based biomimetic delivery strategies display numerous merits, including successive biological functions, innate biocompatibility and superior security since they originate from living organisms, providing a very promising approach. Among them, immune cells receive increasing attention because of their inherent ability in tumor resistance, pathogen elimination, and other significant physiological functions. Herein, we investigated the recent advances on immune cell-based high efficient delivery and therapeutic strategies in solid tumor treatment, mainly focus on T cells, NK cells and macrophages, which have been used as drug cargos directly or provided membrane/exosomes as nanoscale drug delivery systems. We also discuss the further potential applications and perspective of this innovative strategy, as well as the predictable challenges in forward exploration in this emerging area.
Collapse
Affiliation(s)
- Zhuqing Sun
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, CHINA
| | - Jingtong Chen
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, CHINA
| | - Guangcun Chen
- Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou, Jiangsu, 215123, CHINA
| | - Can Zhang
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, CHINA
| | - Chunyan Li
- Suzhou Institute of Nano-tech and Nano-Bionics Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou, Jiangsu, 215123, CHINA
| |
Collapse
|
27
|
Zhang C, Zhang W, Zhu D, Li Z, Wang Z, Li J, Mei X, Xu W, Cheng K, Zhong B. Nanoparticles functionalized with stem cell secretome and CXCR4-overexpressing endothelial membrane for targeted osteoporosis therapy. J Nanobiotechnology 2022; 20:35. [PMID: 35033095 PMCID: PMC8760699 DOI: 10.1186/s12951-021-01231-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/28/2021] [Indexed: 01/16/2023] Open
Abstract
Background Osteoporosis is a chronic condition affecting patients’ morbidity and mortality and represents a big socioeconomic burden. Because stem cells can proliferate and differentiate into bone-forming cells, stem cell therapy for osteoporosis has been widely studied. However, cells as a live drug face multiple challenges because of their instability during preservation and transportation. In addition, cell therapy has potential adverse effects such as embolism, tumorigenicity, and immunogenicity. Results Herein, we sought to use cell-mimicking and targeted therapeutic nanoparticles to replace stem cells. We fabricated nanoparticles (NPs) using polylactic-co-glycolic acid (PLGA) loaded with the secretome (Sec) from mesenchymal stem cells (MSCs) to form MSC-Sec NPs. Furthermore, we cloaked the nanoparticles with the membranes from C–X–C chemokine receptor type 4 (CXCR4)-expressing human microvascular endothelial cells (HMECs) to generate MSC-Sec/CXCR4 NP. CXCR4 can target the nanoparticles to the bone microenvironment under osteoporosis based on the CXCR4/SDF-1 axis. Conclusions In a rat model of osteoporosis, MSC-Sec/CXCR4 NP were found to accumulate in bone, and such treatment inhibited osteoclast differentiation while promoting osteogenic proliferation. In addition, our results showed that MSC-Sec/CXCR4 NPs reduce OVX-induced bone mass attenuation in OVX rats. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01231-6.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.,Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Wei Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Dashuai Zhu
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Zhenhua Li
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Zhenzhen Wang
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Junlang Li
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Xuan Mei
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Wei Xu
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200336, China.
| | - Ke Cheng
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA.
| | - Biao Zhong
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
28
|
Liu Y, Niu H, Wang C, Yang X, Li W, Zhang Y, Ma X, Xu Y, Zheng P, Wang J, Dai K. Bio-inspired, bio-degradable adenosine 5′-diphosphate-modified hyaluronic acid coordinated hydrophobic undecanal-modified chitosan for hemostasis and wound healing. Bioact Mater 2022; 17:162-177. [PMID: 35386451 PMCID: PMC8965034 DOI: 10.1016/j.bioactmat.2022.01.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 02/06/2023] Open
Abstract
Uncontrolled hemorrhage and wound infection are crucial causes of trauma-associated death in both the military and the clinic. Therefore, developing an efficient and rapid hemostatic method with biocompatibility, easy degradation, and wound healing is of great importance and desirability. Inspired by spontaneous blood cell plug formation in the hemostasis process, an adenosine 5′-diphosphate modified pro-coagulation hyaluronic acid (HA-ADP) coordinated with enhanced antibacterial activity of undecanal-modified chitosan (UCS) was fabricated through physical electrostatic cross-linking and freeze-drying. The as-prepared hydrogel sponges showed a porous structure suitable for blood cell adhesion. In particular, the hydrogel exhibited excellent antibacterial ability and promoted the adhesion of platelets and red blood cells, thus inducing a prominent pro-coagulation ability via platelet activation, which exhibits a shorter hemostasis time (58.94% of control) in vitro. Compared with commercially available CELOX and gelatin sponge (GS), HA-ADP/UCS accelerates hemostasis and reduces blood loss in both rat tail amputation and rat artery injury models. Furthermore, all the samples exhibited superior cytocompatibility and biodegradability. Due to these performances, HA-ADP/UCS promoted full-thickness skin defect healing significantly in vivo. All the properties of HA-ADP/UCS suggest that it has great potential for translation as a clinical application material for hemostatic and wound healing. Using adenosine 5'-diphosphate, a physiologically platelet agonist, to modify hyaluronic acids to promote hemostatic effect. Using undecanal to modify Chitosan fabricated with HA-ADP via electrostatic interactions and noncovalent crosslinking method. The hydrogel sponges have excellent antibacterial properties related to the bacterial disruption abilities of UCS. HA-ADP/UCS posed a great hemostatic performance, promoting wound healing by regulating inflammatory response in early stage.
Collapse
Affiliation(s)
- Yihao Liu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Haoyi Niu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
- Corresponding author
| | - Chengwei Wang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
- Corresponding author
| | - Xiaoxiao Yang
- Department of Rehabilitation, Huashan Hospital, Fudan University, No.12 Middle Urumqi Rd, Shanghai, 200040, China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Yuxin Zhang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Xiaojun Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 85 Wujin Rd, Shanghai, 200080, China
| | - Yuanjing Xu
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1956 Huashan Rd, Shanghai, 200030, China
| | - Pengfei Zheng
- Department of Orthopaedic Surgery, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Rd, Nanjing, 210008, China
- Corresponding author
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1956 Huashan Rd, Shanghai, 200030, China
- Corresponding authorShanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China.
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1956 Huashan Rd, Shanghai, 200030, China
| |
Collapse
|
29
|
Li T, Chen T, Chen H, Wang Q, Liu Z, Fang L, Wan M, Mao C, Shen J. Engineered Platelet-Based Micro/Nanomotors for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2104912. [PMID: 34741421 DOI: 10.1002/smll.202104912] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/27/2021] [Indexed: 06/13/2023]
Abstract
Engineered platelets (PLT) can bring new possibilities for diseases treatment due to the specific response for a variety of physiological disease environments. However, the deep penetration of engineered PLT in diseased tissues such as tumor is still an important challenge that restricts the therapeutic effect. Herein, the engineered PLT micromotor (PLT@PDA-DOX) is constructed by a universal self-polymerization modification method of dopamine, and the chemotherapeutic drug doxorubicin (DOX) is loaded by both π-π stacking interaction with polydopamine (PDA) and cellular endocytosis of PLT. The experimental results prove that PLT@PDA-DOX can target to tumor site by the specific binding of PLT with cancer cells, and then the secondary PLT-derived microparticles (PMP@PDA-DOX) are released with the activation of PLT@PDA-DOX by tumor microenvironment (TME). Besides, benefiting from the photothermal conversion capability of PDA, PLT@PDA-DOX micromotors and PMP@PDA-DOX nanomotors are driven by near-infrared light to realize deep penetration. And the PLT-based micro/nanomotors with propulsion capability possess good performance for tumor ablating in vitro and in vivo. In consideration of the operability, mildness, universality of this modification method and the good biocompatibility of PDA, this work may provide a general paradigm for the construction of engineered cells in disease treatment.
Collapse
Affiliation(s)
- Ting Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Tiantian Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Huan Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Qi Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Zhiyong Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Leyi Fang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
30
|
Pan H, Zheng M, Ma A, Liu L, Cai L. Cell/Bacteria-Based Bioactive Materials for Cancer Immune Modulation and Precision Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2100241. [PMID: 34121236 DOI: 10.1002/adma.202100241] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/24/2021] [Indexed: 06/12/2023]
Abstract
Numerous clinical trials for cancer precision medicine research are limited due to the drug resistance, side effects, and low efficacy. Unsatisfactory outcomes are often caused by complex physiologic barriers and abnormal immune events in tumors, such as tumor target alterations and immunosuppression. Cell/bacteria-derived materials with unique bioactive properties have emerged as attractive tools for personalized therapy in cancer. Naturally derived bioactive materials, such as cell and bacterial therapeutic agents with native tropism or good biocompatibility, can precisely target tumors and effectively modulate immune microenvironments to inhibit tumors. Here, the recent advances in the development of cell/bacteria-based bioactive materials for immune modulation and precision therapy in cancer are summarized. Cell/bacterial constituents, including cell membranes, bacterial vesicles, and other active substances have inherited their unique targeting properties and antitumor capabilities. Strategies for engineering living cell/bacteria to overcome complex biological barriers and immunosuppression to promote antitumor efficacy are also summarized. Moreover, past and ongoing trials involving personalized bioactive materials and promising agents such as cell/bacteria-based micro/nano-biorobotics are further discussed, which may become another powerful tool for treatment in the near future.
Collapse
Affiliation(s)
- Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mingbin Zheng
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518112, P. R. China
| | - Aiqing Ma
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
31
|
Ang MJY, Chan SY, Goh YY, Luo Z, Lau JW, Liu X. Emerging strategies in developing multifunctional nanomaterials for cancer nanotheranostics. Adv Drug Deliv Rev 2021; 178:113907. [PMID: 34371084 DOI: 10.1016/j.addr.2021.113907] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer involves a collection of diseases with a common trait - dysregulation in cell proliferation. At present, traditional therapeutic strategies against cancer have limitations in tackling various tumors in clinical settings. These include chemotherapeutic resistance and the inability to overcome intrinsic physiological barriers to drug delivery. Nanomaterials have presented promising strategies for tumor treatment in recent years. Nanotheranostics combine therapeutic and bioimaging functionalities at the single nanoparticle level and have experienced tremendous growth over the past few years. This review highlights recent developments of advanced nanomaterials and nanotheranostics in three main directions: stimulus-responsive nanomaterials, nanocarriers targeting the tumor microenvironment, and emerging nanomaterials that integrate with phototherapies and immunotherapies. We also discuss the cytotoxicity and outlook of next-generation nanomaterials towards clinical implementation.
Collapse
Affiliation(s)
- Melgious Jin Yan Ang
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Siew Yin Chan
- Institute of Materials Research and Engineering, Agency for Science, Technology, and Research, Singapore 138634, Singapore
| | - Yi-Yiing Goh
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jun Wei Lau
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School (ISEP), National University of Singapore, Singapore 119077, Singapore.
| |
Collapse
|
32
|
Bhandarkar S, Prabhakar B, Shende P. Quercetin-loaded platelets as a potential targeted therapy for glioblastoma multiforme cell line U373-MG. Biotechnol J 2021; 16:e2100271. [PMID: 34562072 DOI: 10.1002/biot.202100271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/12/2022]
Abstract
Over the globe, the incidence of glioblastoma multiforme (GM) is very low, that is, 1-4 cases per 100,000, but it is fatal and cancer grows very fast inside the brain tissues, namely astrocytes and oligodendrocytes. Because of the rapid growth, it is difficult to halt the dissemination of tumor in adjacent tissues. Although temozolomide (TMZ) is a currently approved standard of care, it develops resistance over the period. Therefore, there is a need to develop a novel drug delivery system. In this work, authors have developed platelets as drug delivery carriers-loaded with quercetin (QCT) for targeting GM. The effect of QCT and QCT-platelet was assessed on the U373-MG cell line. Natural human platelets were used as carriers for drug loading and drug delivery. Platelets possess an open canalicular system that allows the uptake of drug molecules in the platelet cytoplasm. The study showed that the maximum encapsulation efficiency of QCT-platelet was 93.96 ± 0.12% and the maximum drug release in 24 h was 76.26 ± 0.13% in-vitro at pH 5.5 that mimics the tumor microenvironment. In this work, there is a three-fold enhancement of solubility of QCT. The cytotoxic activity of QCT-platelets was studied in the U373-MG human astrocytoma glioblastoma cell line and the cell viability was 14.52 ± 1.53% after 48 h. Thus, platelets were proved as good carriers for therapeutic moieties and can be effectively used to target the glioblastoma tumor in the near future.
Collapse
Affiliation(s)
- Sayali Bhandarkar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, India
| | - Bala Prabhakar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, India
| |
Collapse
|
33
|
Deng L, Zhang H, Zhang Y, Luo S, Du Z, Lin Q, Zhang Z, Zhang L. An exosome-mimicking membrane hybrid nanoplatform for targeted treatment toward Kras-mutant pancreatic carcinoma. Biomater Sci 2021; 9:5599-5611. [PMID: 34250995 DOI: 10.1039/d1bm00446h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pancreatic carcinoma elevates quickly and thus has a high mortality rate. Therefore, early treatment is essential for treating pancreatic carcinoma. KRAS is the most frequently identified and one of the earliest mutations in pancreatic tumorigenesis. Thus, the KRAS-mutant cell is an ideal target for the treatment of pancreatic carcinoma, especially at the early stage. KRAS mutation increases macropinocytosis in pancreatic cancer cells, enhancing the internalization of exosomes. Because acquiring natural exosomes could be laborious and their encapsulation efficiency is often unsatisfactory, we aimed to develop a delivery system that mimics the Kras-mutant cell targeting capability of exosomes but is easier to generate and has better loading efficiency. For this purpose, we constructed a hybrid nanoplatform by fusing CLT (Celastrol)-Loaded PEGylated lipids with the DC2.4 cell membrane (M-LIP-CLT) to achieve targeted treatment of Kras-mutant pancreatic cancer. This hybrid nanoplatform improved CLT tumor accumulation and showed excellent anti-cancer efficiency both in vitro and in vivo with increased safety. These results suggest that M-LIP-CLT is an effective drug delivery system for targeted therapy against pancreatic carcinoma, and the fusion strategy showed attractive potential for further development.
Collapse
Affiliation(s)
- Lang Deng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Hanming Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Yu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Shi Luo
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Zhengwu Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| |
Collapse
|
34
|
Zhang R, Wu S, Ding Q, Fan Q, Dai Y, Guo S, Ye Y, Li C, Zhou M. Recent advances in cell membrane-camouflaged nanoparticles for inflammation therapy. Drug Deliv 2021; 28:1109-1119. [PMID: 34121563 PMCID: PMC8205088 DOI: 10.1080/10717544.2021.1934188] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
During inflammation, inflammatory cells are rapidly recruited to sites of infection or injury, where they cross physiological barriers around the infected site and further infiltrate into the tissues. Other cells, such as erythrocytes, endothelial cells and stem cells, also play prominent roles in host defense and tissue repair. In recent years, nanotechnology has been exploited to deliver drugs to sites of inflammation. For example, nanoparticles camouflaged with a cell membrane are a novel drug-delivery platform that can interact with the immune system and that show great potential for treating inflammation. Encapsulating drugs inside plasma membranes derived from various cells involved in inflammatory processes can be effective against inflammation. This review describes the preparation, characterization, and properties of various types of cell membrane-camouflaged biomimetic nanoparticles. It also summarizes preclinical research into their efficacy against inflammation.
Collapse
Affiliation(s)
- Rongtao Zhang
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China.,School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqiong Wu
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China.,School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qian Ding
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China.,School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qingze Fan
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Dai
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shiwei Guo
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yun Ye
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Meiling Zhou
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
35
|
Chingale M, Zhu D, Cheng K, Huang K. Bioengineering Technologies for Cardiac Regenerative Medicine. Front Bioeng Biotechnol 2021; 9:681705. [PMID: 34150737 PMCID: PMC8209515 DOI: 10.3389/fbioe.2021.681705] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiac regenerative medicine faces big challenges such as a lack of adult cardiac stem cells, low turnover of mature cardiomyocytes, and difficulty in therapeutic delivery to the injured heart. The interaction of bioengineering and cardiac regenerative medicine offers innovative solutions to this field. For example, cell reprogramming technology has been applied by both direct and indirect routes to generate patient-specific cardiomyocytes. Various viral and non-viral vectors have been utilized for gene editing to intervene gene expression patterns during the cardiac remodeling process. Cell-derived protein factors, exosomes, and miRNAs have been isolated and delivered through engineered particles to overcome many innate limitations of live cell therapy. Protein decoration, antibody modification, and platelet membranes have been used for targeting and precision medicine. Cardiac patches have been used for transferring therapeutics with better retention and integration. Other technologies such as 3D printing and 3D culture have been used to create replaceable cardiac tissue. In this review, we discuss recent advancements in bioengineering and biotechnologies for cardiac regenerative medicine.
Collapse
Affiliation(s)
- Mira Chingale
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Raleigh, NC, United States
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Raleigh, NC, United States
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
36
|
Wang R, Sha X. Biomimetic Drug Delivery Systems Oriented by Biological Function in Tumor Targeting. Curr Drug Targets 2021; 22:882-895. [PMID: 33459231 DOI: 10.2174/1389450122666210114095859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022]
Abstract
The emergence of nanoscale drug delivery systems provides new opportunities for targeting the delivery of chemotherapeutic drugs and has achieved excellent results. In recent years, with the rise in the concept of intelligent drug delivery systems, the design and preparation of carriers have become more and more complicated, which is not conducive to clinical transformation. Researchers are gradually focused on biomimetic nanoscale drug delivery systems, trying to combine the physicochemical properties of nanoscale carriers with the natural biological functions of endogenous substances, so as to boost tumor targeting delivery. In this article, we first classify and introduce biomimetic nanoscale drug delivery systems, and then emphasize their unique biological functions. The biomimetic nanoscale drug delivery systems have the advantages of simple preparation, powerful functions, and low immunogenicity, having a good application prospect.
Collapse
Affiliation(s)
- Rui Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Meng Y, Sun J, Zheng Y, Zhang G, Yu T, Piao H. Platelets: The Emerging Clinical Diagnostics and Therapy Selection of Cancer Liquid Biopsies. Onco Targets Ther 2021; 14:3417-3428. [PMID: 34079287 PMCID: PMC8164876 DOI: 10.2147/ott.s311907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/16/2021] [Indexed: 12/11/2022] Open
Abstract
Due to the inherent molecular heterogeneity of metastatic tumours and the dynamic evolution ability of tumour genomes, tumour tissues obtained through biopsy and other methods cannot capture all of the features of tumour genomes. A new diagnostic concept called “liquid biopsy” has received widespread attention in recent years. Liquid biopsy has changed the clinical practice of oncology and is widely used to guide targeted drug utilization, monitor disease progression and track drug resistance. The latest research subject in liquid biopsy is platelets. Platelets originate from multifunctional haematopoietic stem cells in the bone marrow haematopoietic system. They are small cells from the cytoplasm of bone marrow megakaryocytes. Their main physiological functions are to participate in the processes of physiological haemostasis and coagulation. Tumour cells transfer biomolecules (such as RNA) to platelets through direct contact and release of exosomes, which changes the platelet precursor RNA. Under the stimulation of tumour cells and the tumour microenvironment, platelet precursor mRNA is spliced into mature RNA and converted into functional protein to respond to external stimuli, forming tumour-educated platelets (TEPs). The detection of TEPs in the peripheral blood of patients is expected to be used in clinical tumour diagnosis. This emerging liquid biopsy method can replace and supplement the current tumour detection methods. Further research on the role of platelets in tumour diagnosis will help provide a novel theoretical basis for clinical tumour diagnosis.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Jing Sun
- Department of Biobank, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Yang Zheng
- Department of Clinical Laboratory, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning province Cancer Hospital, Shenyang, 110042, People's Republic of China.,Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| |
Collapse
|
38
|
Samal S, Dash P, Dash M. Drug Delivery to the Bone Microenvironment Mediated by Exosomes: An Axiom or Enigma. Int J Nanomedicine 2021; 16:3509-3540. [PMID: 34045855 PMCID: PMC8149288 DOI: 10.2147/ijn.s307843] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing incidence of bone-related disorders is causing a burden on the clinical scenario. Even though bone is one of the tissues that possess tremendous regenerative potential, certain bone anomalies need therapeutic intervention through appropriate delivery of a drug. Among several nanosystems and biologics that offer the potential to contribute towards bone healing, the exosomes from the class of extracellular vesicles are outstanding. Exosomes are extracellular nanovesicles that, apart from the various advantages, are standing out of the crowd for their ability to conduct cellular communication. The internal cargo of the exosomes is leading to its potential use in therapeutics. Exosomes are being unraveled in terms of the mechanism as well as application in targeting various diseases and tissues. Through this review, we have tried to understand and review all that is already established and the gap areas that still exist in utilizing them as drug delivery vehicles targeting the bone. The review highlights the potential of the exosomes towards their contribution to the drug delivery scenario in the bone microenvironment. A comparison of the pros and cons of exosomes with other prevalent drug delivery systems is also done. A section on the patents that have been generated so far from this field is included.
Collapse
Affiliation(s)
- Sasmita Samal
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha, 751024, India
| | - Pratigyan Dash
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha, 751024, India
| | - Mamoni Dash
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| |
Collapse
|
39
|
Li Q, Song Y, Wang Q, Chen J, Gao J, Tan H, Li S, Wu Y, Yang H, Huang H, Yu Y, Li Y, Zhang N, Huang Z, Pang Z, Qian J, Ge J. Engineering extracellular vesicles with platelet membranes fusion enhanced targeted therapeutic angiogenesis in a mouse model of myocardial ischemia reperfusion. Am J Cancer Res 2021; 11:3916-3931. [PMID: 33664870 PMCID: PMC7914364 DOI: 10.7150/thno.52496] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/09/2021] [Indexed: 01/01/2023] Open
Abstract
Therapeutic angiogenesis is one promising strategy for the treatment of ischemic heart disease, which is the leading cause of death globally. In recent years, extracellular vesicles (EVs) have quickly gained much attention as a cell-free approach to stimulate angiogenesis. However, clinical applications of EVs are limited by their insufficient targeting capability. Herein, we introduce a method to enhance therapeutic angiogenesis based on platelet membrane-engineered EVs. Methods: Platelet-mimetic EVs (P-EVs) were fabricated by fusing the membranes of EVs with platelet membranes by extrusion. A mouse model of myocardial ischemia reperfusion (MI/R) was established and injected with PBS, EVs, and P-EVs to evaluate their targeting ability and therapeutic angiogenesis efficacy. Results: P-EVs inherited the adhesive proteins and natural targeting ability to injured vasculature of platelets and retained the pro-angiogenic potential of EVs. In the MI/R model, P-EVs preferentially accumulated in the injured endothelium of the ischemic hearts and enhanced the angiogenesis potency of EVs. Conclusions: This engineering strategy to modify pre-isolated EVs with platelet membranes by membrane fusion bestows EVs with the targeting ability of platelets and offers an exciting opportunity to design other targeted EVs fused with cell membranes from different sources for therapeutic angiogenesis.
Collapse
|
40
|
Ye S, Liu Y, Lu Y, Ji Y, Mei L, Yang M, Gong X, Gu Q, Li D, Yang F, Li CJ. Cyclic RGD functionalized liposomes targeted to activated platelets for thrombosis dual-mode magnetic resonance imaging. J Mater Chem B 2021; 8:447-453. [PMID: 31833530 DOI: 10.1039/c9tb01834d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Thrombotic disease is a serious threat to human health. The rapid and accurate detection of thrombosis is still a clinical challenge. To achieve the accurate diagnosis of thrombosis with magnetic resonance imaging (MRI), nanomaterials-based contrast agents have been developed in recent years. In this study, cyclic RGD functionalized liposomes targeted to the activated platelets are developed for thrombosis dual-mode MRI. The cyclic RGD functionalized liposomes (cRGD@MLP-Gd) encapsulated with gadolinium diethylenetriamine penta-acetic acid (Gd-DTPA) and superparamagnetic iron oxide (SPIO) are prepared, and their thrombus-targeted T1 and T2 MRI potential is evaluated in vitro and in vivo. Results show that cRGD@MLP-Gd could actively bind to the activated platelets and gradually accumulate at the thrombus site with a T1 - T2 contrast enhancement imaging effect in vitro. In in vivo MRI experiments, cRGD@MLP-Gd exhibits a T2 contrast enhancement at 1 h after intravenous administration, followed by a visibly larger T1 contrast enhancement at the thrombus site. This dynamic property showed that cRGD@MLP-Gd could actively bind to thrombus and possessed an enhanced T1 and T2 dual-mode MRI effect in vivo. Our results establish the characterization, feasibility and superiority of cRGD@MLP-Gd for the rapid identification of thrombosis, showing great potential to improve diagnostic accuracy and sensitivity to thrombosis of the MRI technique.
Collapse
Affiliation(s)
- Sen Ye
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Li L, Fu J, Wang X, Chen Q, Zhang W, Cao Y, Ran H. Biomimetic "Nanoplatelets" as a Targeted Drug Delivery Platform for Breast Cancer Theranostics. ACS APPLIED MATERIALS & INTERFACES 2021; 13:3605-3621. [PMID: 33449625 DOI: 10.1021/acsami.0c19259] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Breast cancer is a major threat to health and lives of females. Biomimetic nanotechnology brought brighter hope for early diagnosis and treatment of breast cancer. Here, we proposed a platelet (PLT) membrane-derived strategy for enhanced photoacoustic (PA)/ultrasonic (US)/fluorescence (FL) multimodal imaging and augmented synergistic photothermal/chemotherapeutic efficacy in tumor cells. A PA imaging contrast and photothermal agent, nanocarbons (CNs), a chemotherapeutic and FL material, doxorubicin (DOX), and perfluoropentane (PFP) were coencapsulated into the poly(lactic-co-glycolic) acid (PLGA) skeletons. Then, the PLT membranes were coated onto the PLGA NPs, which were named as "nanoplatelets" (DOX-PFP-CNs@PLGA/PM NPs). The "nanoplatelets", which conserved the structural advantages and inherent properties of PLTs, could not only escape from phagocytosis of macrophages but also actively targeted tumor cells by the way of antigen-antibody interactions between P-selectin on the PM and CD44 receptors of the tumor cells. With CNs and DOX loaded in, these "nanoplatelets" could serve as an excellent contrast agent for PA/FL imaging. Under laser irradiation, the "nanoplatelets" could turn light energy into heat energy. The laser-triggered photothermal effect, on the one hand, could ablate the tumor cells immediately, and on the other hand, could initiate the optical droplet vaporization of PFP, which subsequently enhanced US imaging and promoted the discharge of encapsulated DOX from the "nanoplatelets" for remarkably strengthening photothermal therapeutic power in turn. In this work, as compared with the bare drug-loaded nanoparticles, the "nanoplatelets" exhibited much more accumulation in the tumor cells, demonstrating superior multimodal imaging capability and preferable synergistic therapeutic performance. In conclusion, the "nanoplatelets" could serve as contrast agents for US imaging and PA imaging to guide the therapy. What is more, the bioinspired PLT-derived, targeted, and nontoxic "nanoplatelets", which were exploited for multimodal PA/US/FL imaging-guided synergistic photothermal/chemo therapy, will be of great value to breast cancer theranostics in the days to come.
Collapse
Affiliation(s)
- Lin Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Jian Fu
- Vascular Surgery Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Xingyue Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Qiaoqi Chen
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Wei Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| |
Collapse
|
42
|
Luo GF, Chen WH, Zeng X, Zhang XZ. Cell primitive-based biomimetic functional materials for enhanced cancer therapy. Chem Soc Rev 2021; 50:945-985. [PMID: 33226037 DOI: 10.1039/d0cs00152j] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cell primitive-based functional materials that combine the advantages of natural substances and nanotechnology have emerged as attractive therapeutic agents for cancer therapy. Cell primitives are characterized by distinctive biological functions, such as long-term circulation, tumor specific targeting, immune modulation etc. Moreover, synthetic nanomaterials featuring unique physical/chemical properties have been widely used as effective drug delivery vehicles or anticancer agents to treat cancer. The combination of these two kinds of materials will catalyze the generation of innovative biomaterials with multiple functions, high biocompatibility and negligible immunogenicity for precise cancer therapy. In this review, we summarize the most recent advances in the development of cell primitive-based functional materials for cancer therapy. Different cell primitives, including bacteria, phages, cells, cell membranes, and other bioactive substances are introduced with their unique bioactive functions, and strategies in combining with synthetic materials, especially nanoparticulate systems, for the construction of function-enhanced biomaterials are also summarized. Furthermore, foreseeable challenges and future perspectives are also included for the future research direction in this field.
Collapse
Affiliation(s)
- Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | | | | | | |
Collapse
|
43
|
Abstract
Exosomes are membrane-bound nano-vehicles shed by most eukaryotic cells. Exosomes contain specific proteins and RNAs from parent cells, and they play key signaling roles in cellular development, modulation, and tissue regeneration. Attempts to isolate and modify exosomes to increase their targeting efficiency to specific tissue are still in their infancy. Here, we describe generation of exosomes from biopsy, isolation of exosomes by centrifugal ultrafiltration method, and approaches for manipulation of cardiac homing exosomes by chemical engineering for the treatment of myocardial infarction.
Collapse
|
44
|
Pei W, Huang B, Chen S, Wang L, Xu Y, Niu C. Platelet-Mimicking Drug Delivery Nanoparticles for Enhanced Chemo-Photothermal Therapy of Breast Cancer. Int J Nanomedicine 2020; 15:10151-10167. [PMID: 33363371 PMCID: PMC7754093 DOI: 10.2147/ijn.s285952] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Traditional nanoparticle-based drug delivery systems suffer from several limitations, such as easy clearance from blood and inaccurate targeting. MATERIALS AND METHODS Here, we developed platelet membrane-coated nanoparticles (PM-NPs) to improve the precise delivery of drugs to tumor sites and enable a more efficient photothermal therapy (PTT) treatment. RESULTS Mimicking the natural platelet membrane, nanoparticles containing drugs and photothermal agents were not recognized and cleared by the immune system; they could circulate in the blood for a long time and accumulate more efficiently at the tumor site, thus releasing more antitumor drugs and achieving better PTT effects. It is worth mentioning that, in this study, we found that tumors in mice treated with the platelet-mimicking nanoparticles were completely eliminated without recurrence during the observation period (up to 18 days). CONCLUSION This study provides a new strategy to design delivery systems of drugs or photothermal agents, whether in biotherapy or other fields.
Collapse
Affiliation(s)
- Wenjing Pei
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
| | - Biying Huang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
| | - Sijie Chen
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan410008, People’s Republic of China
| | - Yan Xu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, People’s Republic of China
| |
Collapse
|
45
|
Jha A, Nikam AN, Kulkarni S, Mutalik SP, Pandey A, Hegde M, Rao BSS, Mutalik S. Biomimetic nanoarchitecturing: A disguised attack on cancer cells. J Control Release 2020; 329:413-433. [PMID: 33301837 DOI: 10.1016/j.jconrel.2020.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
With the changing face of healthcare, there is a demand for drug delivery systems that have increased efficacy and biocompatibility. Nanotechnology derived drug carrier systems were found to be ideal candidates to meet these demands. Among the vast number of nanosized delivery systems, biomimetic nanoparticles have been researched at length. These nanoparticles mimic cellular functions and are highly biocompatible. They are also able to avoid clearance by the reticuloendothelial system which increases the time spent by them in the systemic circulation. Additionally, their low immunogenicity and targeting ability increase their significance as drug carriers. Based on their core material we have summarized them as biomimetic inorganic nanoparticles, biomimetic polymeric nanoparticles, and biomimetic lipid nanoparticles. The core then may be coated using membranes derived from erythrocytes, cancer cells, leukocytes, stem cells, and other membranes to endow them with biomimetic properties. They can be used for personalized therapy and diagnosis of a large number of diseases, primarily cancer. This review summarizes the various therapeutic approaches using biomimetic nanoparticles along with their applications in the field of cancer imaging, nucleic acid therapy and theranostic properties. A brief overview about toxicity concerns related to these nanoconstructs has been added to provide knowledge about biocompatibility of such nanoparticles.
Collapse
Affiliation(s)
- Adrija Jha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Ajinkya Nitin Nikam
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Sadhana P Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Manasa Hegde
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | | | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India.
| |
Collapse
|
46
|
Dash P, Piras AM, Dash M. Cell membrane coated nanocarriers - an efficient biomimetic platform for targeted therapy. J Control Release 2020; 327:546-570. [DOI: 10.1016/j.jconrel.2020.09.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 01/08/2023]
|
47
|
Masterson CH, McCarthy SD, O'Toole D, Laffey JG. The role of cells and their products in respiratory drug delivery: the past, present, and future. Expert Opin Drug Deliv 2020; 17:1689-1702. [PMID: 32842784 DOI: 10.1080/17425247.2020.1814732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Cell-based delivery systems offer considerable promise as novel and innovative therapeutics to target the respiratory system. These systems consist of cells and/or their extracellular vesicles that deliver their contents, such as anti-microbial peptides, micro RNAs, and even mitochondria to the lung, exerting direct therapeutic effects. AREAS COVERED The purpose of this article is to critically review the status of cell-based therapies in the delivery of therapeutics to the lung, evaluate current progress, and elucidate key challenges to the further development of these novel approaches. An overview as to how these cells and/or their products may be modified to enhance efficacy is given. More complex delivery cell-based systems, including cells or vesicles that are genetically modified to (over)express specific therapeutic products, such as proteins and therapeutic nucleic acids are also discussed. Focus is given to the use of the aerosol route to deliver these products directly into the lung. EXPERT OPINION The use of biological carriers to deliver chemical or biological agents demonstrates great potential in modern medicine. The next generation of drug delivery systems may comprise 'cell-inspired' drug carriers that are entirely synthetic, developed using insights from cell-based therapeutics to overcome limitations of current generation synthetic carriers.
Collapse
Affiliation(s)
- Claire H Masterson
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland , Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway , Galway, Ireland
| | - Sean D McCarthy
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland , Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway , Galway, Ireland
| | - Daniel O'Toole
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland , Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway , Galway, Ireland
| | - John G Laffey
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland , Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway , Galway, Ireland.,Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group , Galway, Ireland
| |
Collapse
|
48
|
Chen HY, Deng J, Wang Y, Wu CQ, Li X, Dai HW. Hybrid cell membrane-coated nanoparticles: A multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomater 2020; 112:1-13. [PMID: 32470527 DOI: 10.1016/j.actbio.2020.05.028] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/06/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Biomimetic nanotechnology through camouflaging synthetic nanoparticles (NPs) with natural cell membranes, which bestows with immune evasion and superior targeting capacity, has been extensively used in drug delivery systems (DDS) over the last decades. These biomimetic NPs not only retain the physicochemical features of the synthetic vehicles but also inherit the cell membranes' intrinsic functionalities. Combined with these benefits, optimized nano-biomimetic DDS allow maximum delivery efficacy. Compared to erythrocyte/cancer single cell membrane, the hybrid cell membrane expressing CD47 membrane protein and self-recognition molecules, from erythrocytes and cancer cells, provides remarkable features to the synthetic vehicles, such as immune evasion, long-term circulation, and homotypic targeting. In this review, we describe the preparation strategies, the camouflaging mechanism, and the antitumor applications of hybrid cell membrane-camouflaged NPs. Moreover, we discuss further modification of the hybrid cell membrane and the surface properties of fusion cellular membranes. Finally, we summarize the primary challenges and opportunities associated with these NPs. STATEMENT OF SIGNIFICANCE: Camouflaging synthetic nanoparticles with hybrid cell membrane has been extensively highlighted in recent years. The resultant biomimetic nanoparticles not only reserve the physicochemical properties of the synthetic nanoparticles but also inherit the biological functions of source cells. Compared with single cell membrane, hybrid cell membrane can endow synthetic nanoparticles with multiple biofunctions derived from the original source cells. To provide a timely review of this rapidly developing subject of research, this paper summarized recent progress on the hybrid cell membrane-camouflaged nanoparticles as drug delivery systems for cancer diagnosis and treatment. In this review, we focused primarily on five different types of hybrid cell membrane-camouflaged nanoparticles with the preparation strategies, the camouflaging mechanism, and the antitumor applications. Moreover, further modification of the hybrid cell membrane was also discussed for isolating effectively circulating tumor cells.
Collapse
|
49
|
Tang S, Zhang F, Gong H, Wei F, Zhuang J, Karshalev E, Esteban-Fernández de Ávila B, Huang C, Zhou Z, Li Z, Yin L, Dong H, Fang RH, Zhang X, Zhang L, Wang J. Enzyme-powered Janus platelet cell robots for active and targeted drug delivery. Sci Robot 2020; 5:5/43/eaba6137. [DOI: 10.1126/scirobotics.aba6137] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/30/2020] [Indexed: 12/14/2022]
Abstract
Transforming natural cells into functional biocompatible robots capable of active movement is expected to enhance the functions of the cells and revolutionize the development of synthetic micromotors. However, present cell-based micromotor systems commonly require the propulsion capabilities of rigid motors, external fields, or harsh conditions, which may compromise biocompatibility and require complex actuation equipment. Here, we report on an endogenous enzyme-powered Janus platelet micromotor (JPL-motor) system prepared by immobilizing urease asymmetrically onto the surface of natural platelet cells. This Janus distribution of urease on platelet cells enables uneven decomposition of urea in biofluids to generate enhanced chemophoretic motion. The cell surface engineering with urease has negligible impact on the functional surface proteins of platelets, and hence, the resulting JPL-motors preserve the intrinsic biofunctionalities of platelets, including effective targeting of cancer cells and bacteria. The efficient propulsion of JPL-motors in the presence of the urea fuel greatly enhances their binding efficiency with these biological targets and improves their therapeutic efficacy when loaded with model anticancer or antibiotic drugs. Overall, asymmetric enzyme immobilization on the platelet surface leads to a biogenic microrobotic system capable of autonomous movement using biological fuel. The ability to impart self-propulsion onto biological cells, such as platelets, and to load these cellular robots with a variety of functional components holds considerable promise for developing multifunctional cell-based micromotors for a variety of biomedical applications.
Collapse
Affiliation(s)
- Songsong Tang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Fangyu Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Hua Gong
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Fanan Wei
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Jia Zhuang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Emil Karshalev
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Chuying Huang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhidong Zhou
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhengxing Li
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Lu Yin
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Haifeng Dong
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Ronnie H. Fang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Joseph Wang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
50
|
Li T, Qin X, Li Y, Shen X, Li S, Yang H, Wu C, Zheng C, Zhu J, You F, Liu Y. Cell Membrane Coated-Biomimetic Nanoplatforms Toward Cancer Theranostics. Front Bioeng Biotechnol 2020; 8:371. [PMID: 32411690 PMCID: PMC7202082 DOI: 10.3389/fbioe.2020.00371] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/03/2020] [Indexed: 12/16/2022] Open
Abstract
Research of nanotechnology for cancer therapy and diagnosis extends beyond drug delivery into the targeted site or surveillance the distribution of nanodrugs in vivo or distinction tumor tissue from normal tissue. To satisfy the clinic needs, nanotheranostic platform should hide the surveillance by immune system and the sequestration by filtration organs (i.e., liver and spleen). Use of biologically derived cellular components in the fabrication of nanoparticles can hide these barriers. In this review, we update the recent progress on cell membrane-coated nanoparticles for cancer theranostics. We hope this review paper can inspire further innovations in biomimetic nanomedicine.
Collapse
Affiliation(s)
- Tingting Li
- Department of Biophysics, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiang Qin
- Department of Biophysics, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yichao Li
- Department of Biophysics, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xue Shen
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Shun Li
- Department of Biophysics, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Yang
- Department of Biophysics, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunhui Wu
- Department of Biophysics, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Chuan Zheng
- Department of Cancer Research, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhu
- Department of Cancer Research, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fengming You
- Department of Cancer Research, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiyao Liu
- Department of Biophysics, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Department of Cancer Research, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|