101
|
Binary Effects of Gynostemma Gold Nanoparticles on Obesity and Inflammation via Downregulation of PPARγ/CEPBα and TNF-α Gene Expression. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092795. [PMID: 35566145 PMCID: PMC9104634 DOI: 10.3390/molecules27092795] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/23/2022] [Accepted: 04/23/2022] [Indexed: 02/07/2023]
Abstract
Nanoscience is a multidisciplinary skill with elucidated nanoscale particles and their advantages in applications to various fields. Owing to their economical synthesis, biocompatible nature, and widespread biomedical and environmental applications, the green synthesis of metal nanoparticles using medicinal plants has become a potential research area in biomedical research and functional food formulations. Gynostemma pentaphyllum (GP) has been extensively used in traditional Chinese medicine to cure several diseases, including diabetes mellitus (DM). This is the first study in which we examined the efficacy of G. pentaphyllum gold nanoparticles (GP-AuNPs) against obesity and related inflammation. GP extract was used as a capping agent to reduce Au2+ to Au0 to form stable gold nanoparticles. The nanoparticles were characterized by using UV–VIS spectroscopy, and TEM images were used to analyze morphology. In contrast, the existence of the functional group was measured using FTIR, and size and shape were examined using XRD analysis. In vitro analysis on GP-AuNPs was nontoxic to RAW 264.7 cells and 3T3-L1 cells up to a specific concentration. It significantly decreased lipid accumulation in 3T3-L1 obese and reduced NO production in Raw 264.7 macrophage cells. The significant adipogenic genes PPARγ and CEPBα and a major pro-inflammatory cytokine TNF-α expression were quantified using RT-PCR. The GP-AuNPs decreased the face of these genes remarkably, revealing the antiadipogenic and anti-inflammatory activity of our synthesized GP-AuNPs. This study represents thorough research on the antiobesity effect of Gynostemma pentaphyllum gold nanoparticles synthesized using a green approach and the efficacy instead of related inflammatory responses.
Collapse
|
102
|
Nanotheranostics for Image-Guided Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14050917. [PMID: 35631503 PMCID: PMC9144228 DOI: 10.3390/pharmaceutics14050917] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
Image-guided nanotheranostics have the potential to represent a new paradigm in the treatment of cancer. Recent developments in modern imaging and nanoparticle design offer an answer to many of the issues associated with conventional chemotherapy, including their indiscriminate side effects and susceptibility to drug resistance. Imaging is one of the tools best poised to enable tailoring of cancer therapies. The field of image-guided nanotheranostics has the potential to harness the precision of modern imaging techniques and use this to direct, dictate, and follow site-specific drug delivery, all of which can be used to further tailor cancer therapies on both the individual and population level. The use of image-guided drug delivery has exploded in preclinical and clinical trials although the clinical translation is incipient. This review will focus on traditional mechanisms of targeted drug delivery in cancer, including the use of molecular targeting, as well as the foundations of designing nanotheranostics, with a focus on current clinical applications of nanotheranostics in cancer. A variety of specially engineered and targeted drug carriers, along with strategies of labeling nanoparticles to endow detectability in different imaging modalities will be reviewed. It will also introduce newer concepts of image-guided drug delivery, which may circumvent many of the issues seen with other techniques. Finally, we will review the current barriers to clinical translation of image-guided nanotheranostics and how these may be overcome.
Collapse
|
103
|
Dhas N, García MC, Kudarha R, Pandey A, Nikam AN, Gopalan D, Fernandes G, Soman S, Kulkarni S, Seetharam RN, Tiwari R, Wairkar S, Pardeshi C, Mutalik S. Advancements in cell membrane camouflaged nanoparticles: A bioinspired platform for cancer therapy. J Control Release 2022; 346:71-97. [PMID: 35439581 DOI: 10.1016/j.jconrel.2022.04.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022]
Abstract
The idea of employing natural cell membranes as a coating medium for nanoparticles (NPs) endows man-made vectors with natural capabilities and benefits. In addition to retaining the physicochemical characteristics of the NPs, the biomimetic NPs also have the functionality of source cell membranes. It has emerged as a promising approach to enhancing the properties of NPs for drug delivery, immune evasion, imaging, cancer-targeting, and phototherapy sensitivity. Several studies have been reported with a multitude of approaches to reengineering the surface of NPs using biological membranes. Owing to their low immunogenicity and intriguing biomimetic properties, cell-membrane-based biohybrid delivery systems have recently gained a lot of interest as therapeutic delivery systems. This review summarises different kinds of biomimetic NPs reported so far, their fabrication aspects, and their application in the biomedical field. Finally, it briefs on the latest advances available in this biohybrid concept.
Collapse
Affiliation(s)
- Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Mónica C García
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Ciudad Universitaria, X5000HUA Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Ciudad Universitaria, X5000HUA Córdoba, Argentina
| | - Ritu Kudarha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ajinkya Nitin Nikam
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Gasper Fernandes
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Soji Soman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Raviraja N Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ruchi Tiwari
- Pranveer Singh Institute of Technology, Kanpur, Uttar Pradesh 209305, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, 400056, India
| | - Chandrakantsing Pardeshi
- R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| |
Collapse
|
104
|
Lu L, Xu Q, Wang J, Wu S, Luo Z, Lu W. Drug Nanocrystals for Active Tumor-Targeted Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14040797. [PMID: 35456631 PMCID: PMC9026472 DOI: 10.3390/pharmaceutics14040797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 12/17/2022] Open
Abstract
Drug nanocrystals, which are comprised of active pharmaceutical ingredients and only a small amount of essential stabilizers, have the ability to improve the solubility, dissolution and bioavailability of poorly water-soluble drugs; in turn, drug nanocrystal technology can be utilized to develop novel formulations of chemotherapeutic drugs. Compared with passive targeting strategy, active tumor-targeted drug delivery, typically enabled by specific targeting ligands or molecules modified onto the surface of nanomedicines, circumvents the weak and heterogeneous enhanced permeability and retention (EPR) effect in human tumors and overcomes the disadvantages of nonspecific drug distribution, high administration dosage and undesired side effects, thereby contributing to improving the efficacy and safety of conventional nanomedicines for chemotherapy. Continuous efforts have been made in the development of active tumor-targeted drug nanocrystals delivery systems in recent years, most of which are encouraging and also enlightening for further investigation and clinical translation.
Collapse
Affiliation(s)
- Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular Non-Coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China
- Correspondence:
| |
Collapse
|
105
|
Yang Y, Yin N, Gu Z, Zhao Y, Liu C, Zhou T, Zhang K, Zhang Z, Liu J, Shi J. Engineered biomimetic drug-delivery systems for ischemic stroke therapy. MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
106
|
Yang C, Ming Y, Zhou K, Hao Y, Hu D, Chu B, He X, Yang Y, Qian Z. Macrophage Membrane-Camouflaged shRNA and Doxorubicin: A pH-Dependent Release System for Melanoma Chemo-Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9768687. [PMID: 35233535 PMCID: PMC8851070 DOI: 10.34133/2022/9768687] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/12/2021] [Indexed: 02/05/2023]
Abstract
Improving the efficacy of melanoma treatment remains an important global challenge. Here, we combined chemotherapy with protein tyrosine phosphatase nonreceptor type 2(Ptpn2) based immunotherapy in an effort to address this challenge. Short-hairpin RNA (shRNA) targeting Ptpn2 was coencapsulated with doxorubicin (DOX) in the cell membrane of M1 macrophages (M1HD@RPR). The prepared nanoparticles (NPs) were effectively phagocytosed by B16F10 cells and M1 macrophages, but not by M0 macrophages. Hence, NP evasion from the reticuloendothelial system (RES) was improved and NP enrichment in tumor sites increased. M1HD@RPR can directly kill tumor cells and stimulate immunogenic cell death (ICD) by DOX and downregulate Ptpn2. It can promote M1 macrophage polarization and dendritic cell maturation and increase the proportion of CD8+ T cells. M1HD@RPR killed and inhibited the growth of primary melanoma and lung metastatic tumor cells without harming the surrounding tissue. These findings establish M1HD@RPR as a safe multifunctional nanoparticle capable of effectively combining chemotherapy and gene immunotherapies against melanoma.
Collapse
Affiliation(s)
- Chengli Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China.,Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, China
| | - Yang Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Kai Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Ying Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Danrong Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Bingyang Chu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Yun Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| |
Collapse
|
107
|
Xin T, Cheng L, Zhou C, Zhao Y, Hu Z, Wu X. In-Vivo Induced CAR-T Cell for the Potential Breakthrough to Overcome the Barriers of Current CAR-T Cell Therapy. Front Oncol 2022; 12:809754. [PMID: 35223491 PMCID: PMC8866962 DOI: 10.3389/fonc.2022.809754] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T cell) therapy has shown impressive success in the treatment of hematological malignancies, but the systemic toxicity and complex manufacturing process of current autologous CAR-T cell therapy hinder its broader applications. Universal CAR-T cells have been developed to simplify the production process through isolation and editing of allogeneic T cells from healthy persons, but the allogeneic CAR-T cells have recently encountered safety concerns, and clinical trials have been halted by the FDA. Thus, there is an urgent need to seek new ways to overcome the barriers of current CAR-T cell therapy. In-vivo CAR-T cells induced by nanocarriers loaded with CAR-genes and gene-editing tools have shown efficiency for regressing leukemia and reducing systemic toxicity in a mouse model. The in-situ programming of autologous T-cells avoids the safety concerns of allogeneic T cells, and the manufacture of nanocarriers can be easily standardized. Therefore, the in-vivo induced CAR-T cells can potentially overcome the abovementioned limitations of current CAR-T cell therapy. Here, we provide a review on CAR structures, gene-editing tools, and gene delivery techniques applied in immunotherapy to help design and develop new in-vivo induced CAR-T cells.
Collapse
Affiliation(s)
- Tianqing Xin
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li Cheng
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chuchao Zhou
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yimeng Zhao
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenhua Hu
- Department of Health and Nursing, Nanfang College of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Wu
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
108
|
Zhang Z, Zhou J, Liu C, Zhang J, Shibata Y, Kong N, Corbo C, Harris MB, Tao W. Emerging biomimetic nanotechnology in orthopedic diseases: progress, challenges, and opportunities. TRENDS IN CHEMISTRY 2022. [DOI: 10.1016/j.trechm.2022.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
109
|
Yu S, Wang Y, He P, Shao B, Liu F, Xiang Z, Yang T, Zeng Y, He T, Ma J, Wang X, Liu L. Effective Combinations of Immunotherapy and Radiotherapy for Cancer Treatment. Front Oncol 2022; 12:809304. [PMID: 35198442 PMCID: PMC8858950 DOI: 10.3389/fonc.2022.809304] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Though single tumor immunotherapy and radiotherapy have significantly improved the survival rate of tumor patients, there are certain limitations in overcoming tumor metastasis, recurrence, and reducing side effects. Therefore, it is urgent to explore new tumor treatment methods. The new combination of radiotherapy and immunotherapy shows promise in improving therapeutic efficacy and reducing recurrence by enhancing the ability of the immune system to recognize and eradicate tumor cells, to overcome tumor immune tolerance mechanisms. Nanomaterials, as new drug-delivery-system materials of the 21st century, can maintain the activity of drugs, improve drug targeting, and reduce side effects in tumor immunotherapy. Additionally, nanomaterials, as radiosensitizers, have shown great potential in tumor radiotherapy due to their unique properties, such as light, heat, electromagnetic effects. Here, we review the mechanisms of tumor immunotherapy and radiotherapy and the synergy of radiotherapy with multiple types of immunotherapies, including immune checkpoint inhibitors (ICIs), tumor vaccines, adoptive cell therapy, and cytokine therapy. Finally, we propose the potential for nanomaterials in tumor radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Siting Yu
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ping He
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Bianfei Shao
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Liu
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongzheng Xiang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Yang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Zeng
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao He
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiachun Ma
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiran Wang
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Liu
- Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Lei Liu,
| |
Collapse
|
110
|
Nehru S, Misra R, Bhaswant M. Multifaceted Engineered Biomimetic Nanorobots Toward Cancer Management. ACS Biomater Sci Eng 2022; 8:444-459. [PMID: 35118865 DOI: 10.1021/acsbiomaterials.1c01352] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The noteworthy beneficiary to date in nanotechnology is cancer management. Nanorobots are developed as the result of advancements in the nanostructure, robotics, healthcare, and computer systems. These devices at the nanoscale level are beneficial in the prevention, diagnosis, and treatment of various health conditions notably cancer. Though these structures have distinct potentialities, the usage of inorganic substances in their construction can affect their performance and can cause health issues in the body. To overcome this, naturally inspired substances are incorporated in the fabrication process of nanorobots termed biomimetic nanorobots that can overcome the immunological responses and reduce the side effects with effective functionalization. These biomimetic nanorobots can widen the opportunities in cancer imaging and therapy. Herein, an up-to-date review of biomimetic nanorobots along with their applications in cancer management is provided. Furthermore, the safety issues and future directions of biomimetic nanorobots to achieve clinical translation are also stated.
Collapse
Affiliation(s)
- Sushmitha Nehru
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai-600119, India
| | - Ranjita Misra
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai-600119, India
| | - Maharshi Bhaswant
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai-600119, India
| |
Collapse
|
111
|
Zeng Y, Li S, Zhang S, Wang L, Yuan H, Hu F. Cell membrane coated-nanoparticles for cancer immunotherapy. Acta Pharm Sin B 2022; 12:3233-3254. [PMID: 35967284 PMCID: PMC9366230 DOI: 10.1016/j.apsb.2022.02.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/14/2021] [Accepted: 02/19/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy can effectively inhibit cancer progression by activating the autoimmune system, with low toxicity and high effectiveness. Some of cancer immunotherapy had positive effects on clinical cancer treatment. However, cancer immunotherapy is still restricted by cancer heterogeneity, immune cell disability, tumor immunosuppressive microenvironment and systemic immune toxicity. Cell membrane-coated nanoparticles (CMCNs) inherit abundant source cell-relevant functions, including “self” markers, cross-talking with the immune system, biological targeting, and homing to specific regions. These enable them to possess preferred characteristics, including better biological compatibility, weak immunogenicity, immune escaping, a prolonged circulation, and tumor targeting. Therefore, they are applied to precisely deliver drugs and promote the effect of cancer immunotherapy. In the review, we summarize the latest researches of biomimetic CMCNs for cancer immunotherapy, outline the existing specific cancer immune therapies, explore the unique functions and molecular mechanisms of various cell membrane-coated nanoparticles, and analyze the challenges which CMCNs face in clinical translation.
Collapse
|
112
|
Yang Z, Wang X, Liang G, Yang A, Li J. Photocontrolled chondrogenic differentiation and long-term tracking of mesenchymal stem cells in vivo by upconversion nanoparticles. J Mater Chem B 2022; 10:518-536. [PMID: 34984430 DOI: 10.1039/d1tb02074a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have multiple differentiation potentials and their clinical application is limited by controlled cell differentiation and long-term tracing in vivo. Here, we developed an upconversion nanoparticle (UCNP)-based nanoplatform for the photocontrolled chondrogenic differentiation and long-term tracking of MSCs in vivo. The UCNP nanoplatform could convert 980 nm near-infrared (NIR) light into UV/blue light (365/475 nm) and green/red light (545/647 nm) through Tm/Er doping. Then, the upconverted UV/blue light was used to drive the photosensitive molecule azobenzene (azo) that was modified in mesoporous silica to constantly change its conformation to trigger the release of kartogenin (KGN) from the UCNPs to induce the chondrogeni differentiation of MSCs, achieving photocontrolled cell differentiation. Both in vitro and in vivo experiments demonstrated the effective induction of chondrogenic differentiation in MSCs by NIR light with the UCNP nanoplatform incubation. In addition, after inducing differentiation, the UCNP nanoplatform that remained in the cytoplasm was used as a nanoprobe to monitor the MSCs in vitro and in vivo using the upconverted green/red light under the NIR light. Therefore, the UCNP nanoplatform displayed potential to be a powerful tool for the control of cell differentiation and the simultaneous long-term tracking of MSCs in vivo for regenerative medicine.
Collapse
Affiliation(s)
- Zihan Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China. .,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xichao Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China. .,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Guohai Liang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China. .,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Anli Yang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jinming Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China. .,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
113
|
He R, Zang J, Zhao Y, Dong H, Li Y. Nanotechnology-Based Approaches to Promote Lymph Node Targeted Delivery of Cancer Vaccines. ACS Biomater Sci Eng 2022; 8:406-423. [PMID: 35005881 DOI: 10.1021/acsbiomaterials.1c01274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vaccines are a promising immunotherapy that awakens the human immune system to inhibit and eliminate cancer with fewer side effects compared with traditional radiotherapy and chemotherapy. Although cancer vaccines have shown some efficacy, there are still troublesome bottlenecks to expand their benefits in the clinic, including weak immune effects and limited therapeutic outcomes. In the past few years, in addition to neoantigen screening, a main branch of the efforts has been devoted to promoting the lymph nodes (LNs) targeting of cancer vaccines and the cross-presentation of antigens by dendritic cells (DCs), two cardinal stages in effective initiation of the immune response. Especially, nanomaterials have shown hopeful biomedical applications in the improvement of vaccine effectiveness. This Review briefly outlines the possible mechanisms by which nanoparticle properties affect LN targeting and antigen cross-presentation and then gives an overview of state-of-the-art advances in improving these biological outcomes with nanotechnology.
Collapse
Affiliation(s)
- Ruiqing He
- Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jie Zang
- Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuge Zhao
- Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| | - Haiqing Dong
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yongyong Li
- Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
114
|
Li J, Zhao R, Yang F, Qi X, Ye P, Xie M. An erythrocyte membrance-camouflaged biomimetic nanoplatform for enhanced chemo-photothermal therapy of breast cancer. J Mater Chem B 2022; 10:2047-2056. [DOI: 10.1039/d1tb02522h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nano drug delivery system is a research hotspot in the field of tumor therapy. Molybdenum disulfide (MoS2) nanosheet was selected as the base material and natural red blood cell membrane...
Collapse
|
115
|
Tsai H, Wu Y, Liu X, Xu Z, Liu L, Wang C, Zhang H, Huang Y, Wang L, Zhang W, Su D, Khan FU, Zhu X, Yang R, Pang Y, Eriksson JE, Zhu H, Wang D, Jia B, Cheng F, Chen H. Engineered Small Extracellular Vesicles as a FGL1/PD-L1 Dual-Targeting Delivery System for Alleviating Immune Rejection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102634. [PMID: 34738731 PMCID: PMC8787398 DOI: 10.1002/advs.202102634] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/19/2021] [Indexed: 06/13/2023]
Abstract
There is an urgent need for developing new immunosuppressive agents due to the toxicity of long-term use of broad immunosuppressive agents after organ transplantation. Comprehensive sample analysis revealed dysregulation of FGL1/LAG-3 and PD-L1/PD-1 immune checkpoints in allogeneic heart transplantation mice and clinical kidney transplant patients. In order to enhance these two immunosuppressive signal axes, a bioengineering strategy is developed to simultaneously display FGL1/PD-L1 (FP) on the surface of small extracellular vesicles (sEVs). Among various cell sources, FP sEVs derived from mesenchymal stem cells (MSCs) not only enriches FGL1/PD-L1 expression but also maintain the immunomodulatory properties of unmodified MSC sEVs. Next, it is confirmed that FGL1 and PD-L1 on sEVs are specifically bound to their receptors, LAG-3 and PD-1 on target cells. Importantly, FP sEVs significantly inhibite T cell activation and proliferation in vitro and a heart allograft model. Furthermore, FP sEVs encapsulated with low-dose FK506 (FP sEVs@FK506) exert stronger effects on inhibiting T cell proliferation, reducing CD8+ T cell density and cytokine production in the spleens and heart grafts, inducing regulatory T cells in lymph nodes, and extending graft survival. Taken together, dual-targeting sEVs have the potential to boost the immune inhibitory signalings in synergy and slow down transplant rejection.
Collapse
Affiliation(s)
- Hsiang‐i Tsai
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
- Department of Medical ImagingThe Affiliated Hospital of Jiangsu UniversityZhenjiang212001China
| | - Yingyi Wu
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Xiaoyan Liu
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Longshan Liu
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐Sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Changxi Wang
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐Sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Huanxi Zhang
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐Sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Yisheng Huang
- Department of Oral SurgeryStomatological HospitalSouthern Medical UniversityGuangzhouGuangdong510280P. R China
| | - Linglu Wang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Weixian Zhang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Dandan Su
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | | | - Xiaofeng Zhu
- School of Traditional Medicine Materials ResourceGuangdong Pharmaceutical University YunfuGuangdong527322China
| | - Rongya Yang
- Department of DermatologyThe Seventh Medical Center of PLA General HospitalPeking100010China
| | - Yuxin Pang
- School of Traditional Medicine Materials ResourceGuangdong Pharmaceutical University YunfuGuangdong527322China
| | - John E. Eriksson
- Cell BiologyBiosciencesFaculty of Science and EngineeringÅbo Akademi UniversityTurkuFI‐20520Finland
| | - Haitao Zhu
- Department of Medical ImagingThe Affiliated Hospital of Jiangsu UniversityZhenjiang212001China
| | - Dongqing Wang
- Department of Medical ImagingThe Affiliated Hospital of Jiangsu UniversityZhenjiang212001China
| | - Bo Jia
- Department of Oral SurgeryStomatological HospitalSouthern Medical UniversityGuangzhouGuangdong510280P. R China
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhen518107P. R. China
| |
Collapse
|
116
|
Zheng Y, Li M, Weng B, Mao H, Zhao J. Exosome-based delivery nanoplatforms: Next-generation theranostic platforms for breast cancer. Biomater Sci 2022; 10:1607-1625. [DOI: 10.1039/d2bm00062h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Breast cancer is the most frequent type of malignancy, and the leading cause of cancer-related death in women across the globe. Exosomes are naturally derived 50-150 nm nanovesicles with a...
Collapse
|
117
|
Biomimetic platelet membrane-coated Nanoparticles for targeted therapy. Eur J Pharm Biopharm 2022; 172:1-15. [DOI: 10.1016/j.ejpb.2022.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/18/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023]
|
118
|
Wang H, Zang J, Zhao Z, Zhang Q, Chen S. The Advances of Neutrophil-Derived Effective Drug Delivery Systems: A Key Review of Managing Tumors and Inflammation. Int J Nanomedicine 2021; 16:7663-7681. [PMID: 34815670 PMCID: PMC8605828 DOI: 10.2147/ijn.s328705] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
The chimeric trait of recruitment by inflammatory signals endows neutrophils with the functionality of migrating to inflamed tissues, which can be utilized to tailor novel drug delivery systems. In this review, we introduce a mechanism of neutrophil-derived drug delivery systems recruited into inflamed sites and provide insight into tumors and inflammation therapy. In particular, the advantages of neutrophils—their endogenous-derived neutrophil membrane, exosomes as drug carriers for augmented targeting, prolonged circulation, and improved biostability—were concluded. Subsequently, the latest application in the treatment of tumors and inflammation was elaborated upon, followed by a discussion of the future prospects to neutrophil-derived delivery systems. This promising system will provide new therapeutic avenues for the treatment of inflammation and tumors.
Collapse
Affiliation(s)
- Huaiji Wang
- Department of Nephrology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Jie Zang
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Zihan Zhao
- Department of Dermatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qin Zhang
- Department of Nephrology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Shunjie Chen
- Department of Nephrology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
119
|
Guo L, Zhang Y, Al-Jamal KT. Recent progress in nanotechnology-based drug carriers for celastrol delivery. Biomater Sci 2021; 9:6355-6380. [PMID: 34582530 DOI: 10.1039/d1bm00639h] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Celastrol (CLT) is an active ingredient that was initially discovered and extracted from the root of Tripterygium wilfordii. The potential pharmacological activities of CLT in cancer, obesity, and inflammatory, auto-immune, and neurodegenerative diseases have been demonstrated in recent years. However, CLT's clinical application is extremely restricted by its low solubility/permeability, poor bioavailability, and potential off-target toxicity. The advent of nanotechnology provides a solution to improve the oral bioavailability, therapeutic effects or tissue-targeting ability of CLT. This review focuses on the most recent advances, improvements, inventions, and updated literature of various nanocarrier systems for CLT.
Collapse
Affiliation(s)
- Ling Guo
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Dongqing South Road, Huaxi University City, Guiyang, Guizhou 550025, P.R. China.,Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| | - Yongping Zhang
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Dongqing South Road, Huaxi University City, Guiyang, Guizhou 550025, P.R. China.,Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
120
|
Polla Ravi S, Shamiya Y, Chakraborty A, Elias C, Paul A. Biomaterials, biological molecules, and polymers in developing vaccines. Trends Pharmacol Sci 2021; 42:813-828. [PMID: 34454774 DOI: 10.1016/j.tips.2021.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
Vaccines have been used to train the immune system to recognize pathogens, and prevent and treat diseases, such as cancer, for decades. However, there are continuing challenges in their manufacturing, large-scale production, and storage. Some of them also show suboptimal immunogenicity, requiring additional adjuvants and booster doses. As an alternate vaccination strategy, a new class of biomimetic materials with unique functionalities has emerged in recent years. Here, we explore the current bioengineering techniques that make use of hydrogels, modified polymers, cell membranes, self-assembled proteins, virus-like particles (VLPs), and nucleic acids to deliver and develop biomaterial-based vaccines. We also review design principles and key regulatory issues associated with their development. Finally, we critically assess their limitations, explore approaches to overcome these limitations, and discuss potential future applications for clinical translation.
Collapse
Affiliation(s)
- Shruthi Polla Ravi
- School of Biomedical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Yasmeen Shamiya
- Department of Chemistry, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Cynthia Elias
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada; Biologics Manufacturing Centre, The National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Arghya Paul
- School of Biomedical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada; Department of Chemistry, The University of Western Ontario, London, ON, N6A 5B9, Canada; Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON, N6A 5B9, Canada.
| |
Collapse
|
121
|
Fan Y, Hao W, Cui Y, Chen M, Chu X, Yang Y, Wang Y, Gao C. Cancer Cell Membrane-Coated Nanosuspensions for Enhanced Chemotherapeutic Treatment of Glioma. Molecules 2021; 26:5103. [PMID: 34443689 PMCID: PMC8400986 DOI: 10.3390/molecules26165103] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022] Open
Abstract
Effective intracerebral delivery is key for glioma treatment. However, the drug delivery system within the brain is largely limited by its own adverse physical and chemical properties, low targeting efficiency, the blood-brain barrier and the blood-brain tumor barrier. Herein, we developed a simple, safe and efficient biomimetic nanosuspension. The C6 cell membrane (CCM) was utilized to camouflaged the 10-hydroxycamptothecin nanosuspension (HCPT-NS) in order to obtain HCPT-NS/CCM. Through the use of immune escape and homotypic binding of the cancer cell membrane, HCPT-NS/CCM was able to penetrate the blood-brain barrier and target tumors. The HCPT-NS is only comprised of drugs, as well as a small amount of stabilizers that are characterized by a simple preparation method and high drug loading. Similarly, the HCPT-NS/CCM is able to achieve targeted treatment of glioma without any ligand modification, which leads it to be stable and efficient. Cellular uptake and in vivo imaging experiments demonstrated that HCPT-NS/CCM is able to effectively cross the blood-brain barrier and was concentrated at the glioma site due to the natural homing pathway. Our results reveal that the glioma cancer cell membrane is able to promote drug transport into the brain and enter the tumor via a homologous targeting mechanism.
Collapse
Affiliation(s)
- Yueyue Fan
- College of Pharmacy, Henan University, Kaifeng 475000, China;
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (W.H.); (Y.C.); (M.C.); (Y.Y.)
| | - Wenyan Hao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (W.H.); (Y.C.); (M.C.); (Y.Y.)
| | - Yuexin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (W.H.); (Y.C.); (M.C.); (Y.Y.)
| | - Mengyu Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (W.H.); (Y.C.); (M.C.); (Y.Y.)
| | - Xiaoyang Chu
- Department of Stomatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China;
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (W.H.); (Y.C.); (M.C.); (Y.Y.)
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (W.H.); (Y.C.); (M.C.); (Y.Y.)
| | - Chunsheng Gao
- College of Pharmacy, Henan University, Kaifeng 475000, China;
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (W.H.); (Y.C.); (M.C.); (Y.Y.)
| |
Collapse
|
122
|
Wen J, Moloney EB, Canning A, Donohoe E, Ritter T, Wang J, Xiang D, Wu J, Li Y. Synthesized nanoparticles, biomimetic nanoparticles and extracellular vesicles for treatment of autoimmune disease: Comparison and prospect. Pharmacol Res 2021; 172:105833. [PMID: 34418563 DOI: 10.1016/j.phrs.2021.105833] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022]
Abstract
An emerging strategy is needed to treat autoimmune diseases, many of which are chronic with no definitive cure. Current treatments only alleviate symptoms and have many side effects affecting patient quality of life. Recently, nanoparticle drug delivery systems, an emerging method in medicine, has been used to target cells or organs, without damaging normal tissue. This approach has led to fewer side effects, along with a strong immunosuppressive capacity. Therefore, a nanotechnology approach may help to improve the treatment of autoimmune diseases. In this review, we separated nanoparticles into three categories: synthesized nanoparticles, biomimetic nanoparticles, and extracellular vesicles. This review firstly compares the typical mechanism of action of these three nanoparticle categories respectively in terms of active targeting, camouflage effect, and similarity to parent cells. Then their immunomodulation properties are discussed. Finally, the challenges faced by all these nanoparticles are described.
Collapse
Affiliation(s)
- Jing Wen
- Department of Pharmacy, the Third Hospital of Changsha, Changsha, China
| | - Elizabeth B Moloney
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Aoife Canning
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Ellen Donohoe
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Jiemin Wang
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland.
| | - Daxiong Xiang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Yongjiang Li
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
123
|
Zaheer Y, Vorup‐Jensen T, Webster TJ, Ahmed M, Khan WS, Ihsan A. Protein based nanomedicine: Promising therapeutic modalities against inflammatory disorders. NANO SELECT 2021. [DOI: 10.1002/nano.202100214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Yumna Zaheer
- National Institute for Biotechnology and Genetic Engineering College Pakistan Institute of Engineering and Applied Sciences (NIBGE‐C, PIEAS) Faisalabad Punjab 38000 Pakistan
| | - Thomas Vorup‐Jensen
- Department of Biomedicine and Interdisciplinary Nanoscience Center Aarhus University Aarhus Denmark
| | - Thomas J. Webster
- Department of Chemical Engineering Northeastern University Boston Massachusetts USA
| | - Mukhtiar Ahmed
- Chemistry of Interfaces Luleå University of Technology Luleå Sweden
| | - Waheed S. Khan
- National Institute for Biotechnology and Genetic Engineering College Pakistan Institute of Engineering and Applied Sciences (NIBGE‐C, PIEAS) Faisalabad Punjab 38000 Pakistan
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College Pakistan Institute of Engineering and Applied Sciences (NIBGE‐C, PIEAS) Faisalabad Punjab 38000 Pakistan
| |
Collapse
|
124
|
Le QV, Lee J, Lee H, Shim G, Oh YK. Cell membrane-derived vesicles for delivery of therapeutic agents. Acta Pharm Sin B 2021; 11:2096-2113. [PMID: 34522579 PMCID: PMC8424219 DOI: 10.1016/j.apsb.2021.01.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/02/2021] [Accepted: 01/14/2021] [Indexed: 12/18/2022] Open
Abstract
Cell membranes have recently emerged as a new source of materials for molecular delivery systems. Cell membranes have been extruded or sonicated to make nanoscale vesicles. Unlike synthetic lipid or polymeric nanoparticles, cell membrane-derived vesicles have a unique multicomponent feature, comprising lipids, proteins, and carbohydrates. Because cell membrane-derived vesicles contain the intrinsic functionalities and signaling networks of their parent cells, they can overcome various obstacles encountered in vivo. Moreover, the different natural combinations of membranes from various cell sources expand the range of cell membrane-derived vesicles, creating an entirely new category of drug-delivery systems. Cell membrane-derived vesicles can carry therapeutic agents within their interior or can coat the surfaces of drug-loaded core nanoparticles. Cell membranes typically come from single cell sources, including red blood cells, platelets, immune cells, stem cells, and cancer cells. However, recent studies have reported hybrid sources from two different types of cells. This review will summarize approaches for manufacturing cell membrane-derived vesicles and treatment applications of various types of cell membrane-derived drug-delivery systems, and discuss challenges and future directions.
Collapse
Key Words
- Blood cells
- CAR-T, chimeric antigen receptor-engineered T cell
- CRISPR, clustered regularly interspaced short palindromic repeats
- CXCR4, C-X-C chemokine receptor type 4
- Cancer cells
- Cell membrane-derived vesicles
- DC, dendritic cell
- Drug-delivery systems
- Immune cells
- Manufacturing
- Membrane engineering
- NF-κB, nuclear factor kappa B
- NIR, near infrared
- PEG, polyethylene glycol
- PLGA, poly(lactic-co-glycolic acid)
- RBC, red blood cell
- Stem cells
- TCR, T-cell receptor
- TRAIL, tumor necrosis factor-related apoptosis-inducing ligand
Collapse
Affiliation(s)
- Quoc-Viet Le
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hobin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Gayong Shim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
125
|
Shi Y, Lu A, Wang X, Belhadj Z, Wang J, Zhang Q. A review of existing strategies for designing long-acting parenteral formulations: Focus on underlying mechanisms, and future perspectives. Acta Pharm Sin B 2021; 11:2396-2415. [PMID: 34522592 PMCID: PMC8424287 DOI: 10.1016/j.apsb.2021.05.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/03/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
The need for long-term treatments of chronic diseases has motivated the widespread development of long-acting parenteral formulations (LAPFs) with the aim of improving drug pharmacokinetics and therapeutic efficacy. LAPFs have been proven to extend the half-life of therapeutics, as well as to improve patient adherence; consequently, this enhances the outcome of therapy positively. Over past decades, considerable progress has been made in designing effective LAPFs in both preclinical and clinical settings. Here we review the latest advances of LAPFs in preclinical and clinical stages, focusing on the strategies and underlying mechanisms for achieving long acting. Existing strategies are classified into manipulation of in vivo clearance and manipulation of drug release from delivery systems, respectively. And the current challenges and prospects of each strategy are discussed. In addition, we also briefly discuss the design principles of LAPFs and provide future perspectives of the rational design of more effective LAPFs for their further clinical translation.
Collapse
Key Words
- 2′-F, 2′-fluoro
- 2′-O-MOE, 2′-O-(2-methoxyethyl)
- 2′-OMe, 2′-O-methyl
- 3D, three-dimensional
- ART, antiretroviral therapy
- ASO, antisense oligonucleotide
- Biomimetic strategies
- Chemical modification
- DDS, drug delivery systems
- ECM, extracellular matrix
- ENA, ethylene-bridged nucleic acid
- ESC, enhanced stabilization chemistry
- EVA, ethylene vinyl acetate
- Fc/HSA fusion
- FcRn, Fc receptor
- GLP-1, glucagon like peptide-1
- GS, glycine–serine
- HA, hyaluronic acid
- HES, hydroxy-ethyl-starch
- HP, hypoparathyroidism
- HSA, human serum albumin
- Hydrogels
- ISFI, in situ forming implants
- IgG, immunoglobulin G
- Implantable systems
- LAFs, long-acting formulations
- LAPFs, long-acting parenteral formulations
- LNA, locked nucleic acid
- Long-acting
- MNs, microneedles
- Microneedles
- NDS, nanochannel delivery system
- NPs, nanoparticles
- Nanocrystal suspensions
- OA, osteoarthritis
- PCPP-SA, poly(1,3-bis(carboxyphenoxy)propane-co-sebacic-acid)
- PEG, polyethylene glycol
- PM, platelet membrane
- PMPC, poly(2-methyacryloyloxyethyl phosphorylcholine)
- PNAs, peptide nucleic acids
- PS, phase separation
- PSA, polysialic acid
- PTH, parathyroid hormone
- PVA, polyvinyl alcohol
- RBCs, red blood cells
- RES, reticuloendothelial system
- RNAi, RNA interference
- SAR, structure‒activity relationship
- SCID, severe combined immunodeficiency
- SE, solvent extraction
- STC, standard template chemistry
- TNFR2, tumor necrosis factor receptor 2
- hGH, human growth hormone
- im, intramuscular
- iv, intravenous
- mPEG, methoxypolyethylene glycol
- sc, subcutaneous
Collapse
Affiliation(s)
- Yujie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiangyu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
126
|
Artificial cells for the treatment of liver diseases. Acta Biomater 2021; 130:98-114. [PMID: 34126265 DOI: 10.1016/j.actbio.2021.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/06/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022]
Abstract
Liver diseases have become an increasing health burden and account for over 2 million deaths every year globally. Standard therapies including liver transplant and cell therapy offer a promising treatment for liver diseases, but they also suffer limitations such as adverse immune reactions and lack of long-term efficacy. Artificial cells that mimic certain functions of a living cell have emerged as a new strategy to overcome some of the challenges that liver cell therapy faces at present. Artificial cells have demonstrated advantages in long-term storage, targeting capability, and tuneable features. This article provides an overview of the recent progress in developing artificial cells and their potential applications in liver disease treatment. First, the design of artificial cells and their biomimicking functions are summarized. Then, systems that mimic cell surface properties are introduced with two concepts highlighted: cell membrane-coated artificial cells and synthetic lipid-based artificial cells. Next, cell microencapsulation strategy is summarized and discussed. Finally, challenges and future perspectives of artificial cells are outlined. STATEMENT OF SIGNIFICANCE: Liver diseases have become an increasing health burden. Standard therapies including liver transplant and cell therapy offer a promising treatment for liver diseases, but they have limitations such as adverse immune reactions and lack of long-term efficacy. Artificial cells that mimic certain functions of a living cell have emerged as a new strategy to overcome some of the challenges that liver cell therapy faces at present. This article provides an overview of the recent progress in developing artificial cells and their potential applications in liver disease treatment, including the design of artificial cells and their biomimicking functions, two systems that mimic cell surface properties (cell membrane-coated artificial cells and synthetic lipid-based artificial cells), and cell microencapsulation strategy. We also outline the challenges and future perspectives of artificial cells.
Collapse
|
127
|
Liu X, Zhong X, Li C. Challenges in cell membrane-camouflaged drug delivery systems: Development strategies and future prospects. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.03.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
128
|
Bender EC, Kraynak CA, Huang W, Suggs LJ. Cell-Inspired Biomaterials for Modulating Inflammation. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:279-294. [PMID: 33528306 DOI: 10.1089/ten.teb.2020.0276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Inflammation is a crucial part of wound healing and pathogen clearance. However, it can also play a role in exacerbating chronic diseases and cancer progression when not regulated properly. A subset of current innate immune engineering research is focused on how molecules such as lipids, proteins, and nucleic acids native to a healthy inflammatory response can be harnessed in the context of biomaterial design to promote healing, decrease disease severity, and prolong survival. The engineered biomaterials in this review inhibit inflammation by releasing anti-inflammatory cytokines, sequestering proinflammatory cytokines, and promoting phenotype switching of macrophages in chronic inflammatory disease models. Conversely, other biomaterials discussed here promote inflammation by mimicking pathogen invasion to inhibit tumor growth in cancer models. The form that these biomaterials take spans a spectrum from nanoparticles to large-scale hydrogels to surface coatings on medical devices. Cell-inspired molecules have been incorporated in a variety of creative ways, including loaded into or onto the surface of biomaterials or used as the biomaterials themselves.
Collapse
Affiliation(s)
- Elizabeth C Bender
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Chelsea A Kraynak
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Wenbai Huang
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA.,Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas, USA
| | - Laura J Suggs
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
129
|
Chattopadhyay M, Orlikowska H, Krok E, Piatkowski L. Sensing Hydration of Biomimetic Cell Membranes. BIOSENSORS 2021; 11:241. [PMID: 34356712 PMCID: PMC8301980 DOI: 10.3390/bios11070241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 11/17/2022]
Abstract
Biological membranes play a vital role in cell functioning, providing structural integrity, controlling signal transduction, and controlling the transport of various chemical species. Owing to the complex nature of biomembranes, the self-assembly of lipids in aqueous media has been utilized to develop model systems mimicking the lipid bilayer structure, paving the way to elucidate the mechanisms underlying various biological processes, as well as to develop a number of biomedical and technical applications. The hydration properties of lipid bilayers are crucial for their activity in various cellular processes. Of particular interest is the local membrane dehydration, which occurs in membrane fusion events, including neurotransmission, fertilization, and viral entry. The lack of universal technique to evaluate the local hydration state of the membrane components hampers understanding of the molecular-level mechanisms of these processes. Here, we present a new approach to quantify the hydration state of lipid bilayers. It takes advantage of the change in the lateral diffusion of lipids that depends on the number of water molecules hydrating them. Using fluorescence recovery after photobleaching technique, we applied this approach to planar single and multicomponent supported lipid bilayers. The method enables the determination of the hydration level of a biomimetic membrane down to a few water molecules per lipid.
Collapse
Affiliation(s)
| | | | | | - Lukasz Piatkowski
- Faculty of Materials Engineering and Technical Physics, Institute of Physics, Division of Molecular Physics, Poznan University of Technology, Piotrowo 3, 60-965 Poznan, Poland; (H.O.); (E.K.)
| |
Collapse
|
130
|
Direct in vivo reprogramming with non-viral sequential targeting nanoparticles promotes cardiac regeneration. Biomaterials 2021; 276:121028. [PMID: 34293701 DOI: 10.1016/j.biomaterials.2021.121028] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/23/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022]
Abstract
microRNA-mediated direct cardiac reprogramming, directly converts fibroblasts into induced cardiomyocyte-like cells (iCMs), which holds great promise in cardiac regeneration therapy. However, effective approaches to deliver therapeutic microRNA into cardiac fibroblasts (CFs) to induce in vivo cardiac reprogramming remain to be explored. Herein, a non-viral biomimetic system to directly reprogram CFs for cardiac regeneration after myocardial injury was developed by coating FH peptide-modified neutrophil-mimicking membranes on mesoporous silicon nanoparticles (MSNs) loaded with microRNA1, 133, 208, and 499 (miR Combo). Through utilizing the natural inflammation-homing ability of neutrophil membrane protein and FH peptide's high affinity to tenascin-C (TN-C) produced by CFs, this nanoparticle could realize sequential targeting to CFs in the injured heart and precise intracellular delivery of miRCombo, which induced reprogramming resident CFs into iCMs. In a mouse model of myocardial ischemia/reperfusion injury, intravenous injection of the nanoparticles successfully delivered miRCombo into fibroblasts and led to efficient reprogramming, resulting in improved cardiac function and attenuated fibrosis. This delivery system is minimally invasive and bio-safe, providing a proof-of-concept for biomimetic and sequential targeting nanomedicine delivery system for microRNA-mediated reprogramming therapy in multiple diseases.
Collapse
|
131
|
Li A, Zhao Y, Li Y, Jiang L, Gu Y, Liu J. Cell-derived biomimetic nanocarriers for targeted cancer therapy: cell membranes and extracellular vesicles. Drug Deliv 2021; 28:1237-1255. [PMID: 34142930 PMCID: PMC8216268 DOI: 10.1080/10717544.2021.1938757] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanotechnology provides synthetic carriers for cancer drug delivery that protect cargos from degradation, control drug release and increase local accumulation at tumors. However, these non-natural vehicles display poor tumor targeting and potential toxicity and are eliminated by the immune system. Recently, biomimetic nanocarriers have been widely developed based on the concept of ‘mimicking nature.’ Among them, cell-derived biomimetic vehicles have become the focus of bionics research because of their multiple natural functions, such as low immunogenicity, long circulation time and targeting ability. Cell membrane-coated carriers and extracellular vesicles are two widely used cell-based biomimetic materials. Here, this review summarizes the latest progress in the application of these two biomimetic carriers in targeted cancer therapy. Their properties and performance are compared, and their future challenges and development prospects are discussed.
Collapse
Affiliation(s)
- Aixue Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunan Zhao
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yixiu Li
- Department of Pharmacy, Shanghai Integrated Traditional Chinese and Western Medicine Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liangdi Jiang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
132
|
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
|
133
|
Wang R, Zhang Z, Liu B, Xue J, Liu F, Tang T, Liu W, Feng F, Qu W. Strategies for the design of nanoparticles: starting with long-circulating nanoparticles, from lab to clinic. Biomater Sci 2021; 9:3621-3637. [PMID: 34008587 DOI: 10.1039/d0bm02221g] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Short half-life is one of the main causes of drug attrition in clinical development, which also leads to the failure of many leading compounds and hits to become drug candidates. Nowadays, nanomaterials have been applied to drug development to address this problem. In fact, the clinical application of nanoparticles (NPs) is severely limited due to their rapid elimination by the reticuloendothelial system (RES) in vivo. In this paper, we aim to summarize representative strategies on prolonging the circulation time for bridging the gap between excellent pharmaceutics and proper half-life and encourage clinical translation.
Collapse
Affiliation(s)
- Ruyi Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Zhongtao Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Bowen Liu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Jingwei Xue
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Taian City institute of Digestive Disease, Taian City Central Hospital, Taian, 271000, China
| | - Fulei Liu
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Pharmaceutical Department, Taian City Central Hospital, Taian, 271000, China
| | - Tongzhong Tang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China and Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China. and Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, China.
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
134
|
Zhang H, Lu F, Pan W, Ge Y, Cui B, Gong S, Li N, Tang B. A dual-catalytic nanoreactor for synergistic chemodynamic-starvation therapy toward tumor metastasis suppression. Biomater Sci 2021; 9:3814-3820. [PMID: 33881052 DOI: 10.1039/d1bm00240f] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Tumor metastasis is extremely deadly for cancer patients and developing effective treatments for deep metastatic tumors remains a major challenge. In this study, we demonstrated a dual-catalytic nanoreactor for tumor metastasis suppression by synergistic Fenton reaction activated chemodynamic therapy (CDT) and glucose oxidase (GOx) initiated starvation therapy. GOx on the surface of hollow mesoporous silica nanoparticles can catalyze the decomposition of intratumoral glucose to generate gluconic acid and H2O2, while Fe3O4 nanoparticles as a Fenton reaction catalyst can in situ catalyze H2O2 to produce highly toxic hydroxyl radicals (˙OH). The oxygen-carrying perfluorohexane (PFC) in the hole of the hollow structures can alleviate the hypoxic environment and promote dual-catalytic reactions. After being disguised by the cancer cell membrane, the delivery efficiency and biological safety of the nanoreactor were effectively improved. The nanoreactor can realize sequential glucose depletion and ˙OH aggregation, which effectively suppress tumor metastasis with negligible side effects.
Collapse
Affiliation(s)
- Huiwen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Fei Lu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Yegang Ge
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bingjie Cui
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Shaohua Gong
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
135
|
Cell membrane cloaked nanomedicines for bio-imaging and immunotherapy of cancer: Improved pharmacokinetics, cell internalization and anticancer efficacy. J Control Release 2021; 335:130-157. [PMID: 34015400 DOI: 10.1016/j.jconrel.2021.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/13/2023]
Abstract
Despite enormous advancements in the field of oncology, the innocuous and effectual treatment of various types of malignancies remained a colossal challenge. The conventional modalities such as chemotherapy, radiotherapy, and surgery have been remained the most viable options for cancer treatment, but lacking of target-specificity, optimum safety and efficacy, and pharmacokinetic disparities are their impliable shortcomings. Though, in recent decades, numerous encroachments in the field of onco-targeted drug delivery have been adapted but several limitations (i.e., short plasma half-life, early clearance by reticuloendothelial system, immunogenicity, inadequate internalization and localization into the onco-tissues, chemoresistance, and deficient therapeutic efficacy) associated with these onco-targeted delivery systems limits their clinical viability. To abolish the aforementioned inadequacies, a promising approach has been emerged in which stealthing of synthetic nanocarriers has been attained by cloaking them into the natural cell membranes. These biomimetic nanomedicines not only retain characteristics features of the synthetic nanocarriers but also inherit the cell-membrane intrinsic functionalities. In this review, we have summarized preparation methods, mechanism of cloaking, and pharmaceutical and therapeutic superiority of cell-membrane camouflaged nanomedicines in improving the bio-imaging and immunotherapy against various types of malignancies. These pliable adaptations have revolutionized the current drug delivery strategies by optimizing the plasma circulation time, improving the permeation into the cancerous microenvironment, escaping the immune evasion and rapid clearance from the systemic circulation, minimizing the immunogenicity, and enabling the cell-cell communication via cell membrane markers of biomimetic nanomedicines. Moreover, the preeminence of cell-membrane cloaked nanomedicines in improving the bio-imaging and theranostic applications, alone or in combination with phototherapy or radiotherapy, have also been pondered.
Collapse
|
136
|
Chen X, Liu B, Tong R, Zhan L, Yin X, Luo X, Huang Y, Zhang J, He W, Wang Y. Orchestration of biomimetic membrane coating and nanotherapeutics in personalized anticancer therapy. Biomater Sci 2021; 9:590-625. [PMID: 33305765 DOI: 10.1039/d0bm01617a] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nanoparticle-based therapeutic and detectable modalities can augment anticancer efficiency, holding potential in capable target and suppressive metastases post administration. However, the individual discrepancies of the current "one-size-fits-all" strategies for anticancer nanotherapeutics have heralded the need for "personalized therapy". Benefiting from the special inherency of various cells, diverse cell membrane-coated nanoparticles (CMCNs) were established on a patient-by-patient basis, which would facilitate the personalized treatment of individual cancer patients. CMCNs in a complex microenvironment can evade the immune system and target homologous tumors with a suppressed immune response, as well as a prolonged circulation time, consequently increasing the drug accumulation at the tumor site and anticancer therapeutic efficacy. This review focuses on the emerging strategies and advances of CMCNs to synergistically integrate the merit of source cells with nanoparticulate delivery systems for the orchestration of personalized anticancer nanotherapeutics, thus discussing their rationalities in facilitating chemotherapy, imaging, immunotherapy, phototherapy, radiotherapy, sonodynamic, magnetocaloric, chemodynamic and gene therapy. Furthermore, the mechanism, challenges and opportunities of CMCNs in personalized anticancer therapy were highlighted to further boost cooperation from different fields, including materials science, chemistry, medicine, pharmacy and biology for the lab-to-clinic translation of CMCNs combined with the individual advantages of source cells and nanotherapeutics.
Collapse
Affiliation(s)
- Xuerui Chen
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Bingbing Liu
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Rongliang Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lin Zhan
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xuelian Yin
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xin Luo
- Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanan Huang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Wen He
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanli Wang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
137
|
Li X, Wang L, Wang L, Yu J, Lu G, Zhao W, Miao C, Zou C, Wu J. Overcoming therapeutic failure in osteosarcoma via Apatinib-encapsulated hydrophobic poly(ester amide) nanoparticles. Biomater Sci 2021; 8:5888-5899. [PMID: 33001086 DOI: 10.1039/d0bm01296c] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anti-angiogenic tyrosine kinase inhibitors (TKIs) have been proved to be effective in prolonging progression-free survival in advanced osteosarcoma. However, osteosarcoma stem-like cells persist for a long time and ultimately cause disease recurrence and therapy resistance. Here, we reveal that inefficient accumulation of Apatinib, an anti-angiogenic TKI, induces the expression of ribosome-associated genes in osteosarcoma, and confers apoptosis resistance. An engineered nanoscale delivery system based on hydrophobic poly(ester amide) has been established to effectively deliver Apatinib to improve the treatment. Notably, the considerable uptake by osteosarcoma cells enables this nanodrug to distribute increasingly inside the tumor. Furthermore, the delivered nano-Apatinib can suppress osteosarcoma stemness and enhance osteosarcoma stem-like cell apoptosis, and overcomes the crucial bottleneck of the unfavorable stem-like cell residue for TKI therapy. Importantly, nano-Apatinib significantly inhibits the osteosarcoma stem-like cell-derived tumor growth in contrast with free Apatinib, with minimal side effects. These results suggest that this Apatinib-loaded nano delivery system may serve as a promising strategy to solve the issue of TKI therapeutic resistance existing in advanced osteosarcoma.
Collapse
Affiliation(s)
- Xiangyu Li
- The Stem Cell and Tissue Engineering Research Center, Changzhi Medical College, Changzhi, Shanxi 046000, P. R. China.
| | - Liying Wang
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Li Wang
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China
| | - Jiaming Yu
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China
| | - Guohao Lu
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China
| | - Wei Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China
| | - Congxiu Miao
- The Stem Cell and Tissue Engineering Research Center, Changzhi Medical College, Changzhi, Shanxi 046000, P. R. China.
| | - Changye Zou
- Musculoskeletal Oncology Center, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.
| | - Jun Wu
- The Stem Cell and Tissue Engineering Research Center, Changzhi Medical College, Changzhi, Shanxi 046000, P. R. China. and School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou 510006, China.
| |
Collapse
|
138
|
Chen Y, Ma H, Wang W, Zhang M. A size-tunable nanoplatform: enhanced MMP2-activated chemo-photodynamic immunotherapy based on biodegradable mesoporous silica nanoparticles. Biomater Sci 2021; 9:917-929. [PMID: 33284292 DOI: 10.1039/d0bm01452d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although immunotherapy is emerging as a revolutionary strategy for cancer therapy, its clinical effect is severely impaired by adaptive immune evasion and inefficient activation of antitumor immune response. Photodynamic therapy and chemotherapy have been shown to efficiently enhance the therapeutic effect of PD-L1 immunotherapy via different mechanisms. However, the lack of a precise drug delivery system seriously impedes the clinical application of combination therapy. To address these restrictions, a matrix metalloproteinases-2 (MMP2)-activated shrinkable nanosystem was developed to potentiate the antitumor efficacy of anti-PD-L1 antibody (aPDL1) delivered along with a chemo-photodynamic therapy. The nanosystem maintains its structure to accelerate tumor accumulation and shrinks down to a smaller size to facilitate tumor penetration and cellular uptake upon arriving in the tumor microenvironment. The exposure of aPDL1 on the surface of the biodegradable mesoporous silica cores (bMSNs) blocks the PD-1/PD-L1 interaction between tumor cells and T cells. Meanwhile, photosensitizer chlorin e6 (Ce6) and paclitaxel (PTX) loaded bMSNs effectively enter tumor cells and induce chemo-photodynamic therapy. The nanosystem elicits a chemo-photodynamic-induced immune response and improves the therapeutic effect of PD-L1 blockade mediated by aPDL1. Furthermore, the nanosystem displays a sustained prohibitive effect on tumor metastasis to distant sites. Our work presents a promising strategy for enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Ye Chen
- Department of Pharmacy, Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao 266034, China
| | - He Ma
- Institute of Biomedical Materials and Engineering, College of Materials Sciences and Engineering, Qingdao University, Qingdao 266071, China.
| | - Wenli Wang
- Institute of Biomedical Materials and Engineering, College of Materials Sciences and Engineering, Qingdao University, Qingdao 266071, China.
| | - Min Zhang
- Institute of Biomedical Materials and Engineering, College of Materials Sciences and Engineering, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
139
|
Wang H, Zhang C, Zhang Y, Tian R, Cheng G, Pan H, Cui M, Chang J. An efficient delivery of photosensitizers and hypoxic prodrugs for a tumor combination therapy by membrane camouflage nanoparticles. J Mater Chem B 2021; 8:2876-2886. [PMID: 32191252 DOI: 10.1039/d0tb00235f] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Photodynamic therapy (PDT) is an oxygen-dependent, non-invasive cancer treatment. The hypoxia in the tumor environment limits the therapeutic effects of PDT. The combined delivery of photosensitizers and hypoxic prodrugs is expected to improve the efficacy of tumor treatment. In this paper, an erythrocyte and tumor cell membrane camouflage nanocarrier co-loaded with a photosensitizer (indocyanine green) and a hypoxic prodrug (tirapazamine) were used to combine PDT with chemotherapy. The system achieved less macrophage clearance through erythrocyte membranes and tumor-targeted tumor cell membranes, thereby inducing cell death and increasing tumor environment hypoxia by NIR irradiation of photosensitizers. Furthermore, the hypoxic environment activated TPZ to kill more tumor cells. In vivo results showed that the tumor inhibition rate of the drug-loaded nanoparticles increased from 34% to 64% after membrane modification. Moreover, the tumor inhibition rate of the photodynamic treatment group alone was only 47%, and the tumor inhibition rate after the combination was 1.3 times that of photodynamic therapy alone. Our platform is expected to contribute to the further application of cancer combination therapy.
Collapse
Affiliation(s)
- Hanjie Wang
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| | - Chaonan Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| | - Yingying Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| | - Ran Tian
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| | - Guohui Cheng
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| | - Huizhuo Pan
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| | - Meihui Cui
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| | - Jin Chang
- School of Life Sciences, Tianjin University, Tianjin, 300072, P. R. China. and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin, 300072, P. R. China
| |
Collapse
|
140
|
Wei R, Dong Y, Tu Y, Luo S, Pang X, Zhang W, Yao W, Tang W, Yang H, Wei X, Jiang X, Yuan Y, Yang R. Bioorthogonal Pretargeting Strategy for Anchoring Activatable Photosensitizers on Plasma Membranes for Effective Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:14004-14014. [PMID: 33728894 DOI: 10.1021/acsami.1c01259] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Developing novel activatable photosensitizers with excellent plasma membrane targeting ability is urgently needed for smart photodynamic therapy (PDT). Herein, a tumor acidity-activatable photosensitizer combined with a two-step bioorthogonal pretargeting strategy to anchor photosensitizers on the plasma membrane for effective PDT is developed. Briefly, artificial receptors are first anchored on the cell plasma membrane using cell-labeling agents (Az-NPs) via the enhanced permeability and retention effect to achieve the tumor cell labeling. Then, pH-sensitive nanoparticles (S-NPs) modified with dibenzocyclooctyne (DBCO) and chlorin e6 (Ce6) accumulate in tumor tissue and disassemble upon protonation of their tertiary amines in response to the acidic tumor environment, exposing the contained DBCO and Ce6. The selective, highly specific click reactions between DBCO and azide groups enable Ce6 to be anchored on the tumor cell surface. Upon laser irradiation, the cell membrane is severely damaged by the cytotoxic reactive oxygen species, resulting in remarkable cellular apoptosis. Taken together, the membrane-localized PDT by our bioorthogonal pretargeting strategy to anchor activatable photosensitizers on the plasma membrane provides a simple but effective method for enhancing the therapeutic efficacy of photosensitizers in anticancer therapy.
Collapse
Affiliation(s)
- Ruili Wei
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Yansong Dong
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Yalan Tu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
| | - Shiwei Luo
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Xinrui Pang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Wanli Zhang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Wang Yao
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Wenjie Tang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Huikang Yang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Xinqing Jiang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Youyong Yuan
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, P.R. China
| | - Ruimeng Yang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| |
Collapse
|
141
|
Mauri E, Gori M, Giannitelli SM, Zancla A, Mozetic P, Abbruzzese F, Merendino N, Gigli G, Rossi F, Trombetta M, Rainer A. Nano-encapsulation of hydroxytyrosol into formulated nanogels improves therapeutic effects against hepatic steatosis: An in vitro study. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112080. [PMID: 33947572 DOI: 10.1016/j.msec.2021.112080] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/12/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022]
Abstract
Nanomaterials hold promise as a straightforward approach for enhancing the performance of bioactive compounds in several healthcare scenarios. Indeed, nanoencapsulation represents a valuable strategy to preserve the bioactives, maximizing their bioavailability. Here, a nanoencapsulation strategy for the treatment of nonalcoholic fatty liver disease (NAFLD) is presented. NAFLD represents the most common chronic liver disease in Western societies, and still lacks an effective therapy. Hydroxytyrosol (HT), a naturally occurring polyphenol, has been shown to protect against hepatic steatosis through its lipid-lowering, antioxidant and anti-inflammatory activities. However, the efficient delivery of HT to hepatocytes remains a crucial aspect: the design of smart nanogels appears as a promising tool to promote its intracellular uptake. In this paper, we disclose the synthesis of nanogel systems based on polyethylene glycol and polyethyleneimine which have been tested in an in vitro model of hepatic steatosis at two different concentrations (0.1 mg/mL and 0.5 mg/mL), taking advantage of high-content analysis tools. The proposed HT-loaded nanoscaffolds are non-toxic to cells, and their administration showed a significant decrease in the intracellular triglyceride levels, restoring cell viability and outperforming the results achievable with HT in its non-encapsulated form. Moreover, nanogels do not induce oxidative stress, thus demonstrating their biosafety. Overall, the formulated nanogel system achieves superior performance compared to conventional drug administration routes and hence represents a promising strategy for the management of NAFLD.
Collapse
Affiliation(s)
- Emanuele Mauri
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Manuele Gori
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), via E. Ramarini 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Sara Maria Giannitelli
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Andrea Zancla
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy; Department of Engineering, Università degli Studi di Roma Tre, via Vito Volterra 62, 00146 Rome, Italy
| | - Pamela Mozetic
- Institute of Nanotechnology (NANOTEC), National Research Council (CNR), via Monteroni, 73100 Lecce, Italy
| | - Franca Abbruzzese
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Nicolò Merendino
- Department of Ecology and Biology, Università degli Studi della Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology (NANOTEC), National Research Council (CNR), via Monteroni, 73100 Lecce, Italy; Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, via Arnesano, 73100 Lecce, Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, via L. Mancinelli 7, 20131 Milan, Italy
| | - Marcella Trombetta
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128 Rome, Italy; Institute of Nanotechnology (NANOTEC), National Research Council (CNR), via Monteroni, 73100 Lecce, Italy.
| |
Collapse
|
142
|
Zhang Q, Fan A, Fu J, Zhao Q. Precise engineering of iron oxide nanoparticle-encapsulated protein hydrogel: Implications for cardiac toxicity and ultrasound contrast agents. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.01.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
143
|
Platelet membrane camouflaged nanoparticles: Biomimetic architecture for targeted therapy. Int J Pharm 2021; 598:120395. [PMID: 33639226 DOI: 10.1016/j.ijpharm.2021.120395] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/27/2021] [Accepted: 02/12/2021] [Indexed: 12/31/2022]
Abstract
Cell membrane coating strategy is one of the promising techniques for biomimetic functionalization of nanoparticle. The biomimetic nanoparticles camouflage themselves utilizing the fundamental properties of native cells. Cell membranes are extracted from various cells to cloak the nanoparticles for targeted drug delivery. Platelet membrane is one such cell membrane proposing itself as a potential camouflager to escape the immune surveillance and aid prolonged blood circulation with minimum systemic cytotoxicity. Platelets play a very important role in the physiological functions of the body and also feature in few pathological disorders like cancer, atherosclerosis and rheumatoid arthritis. This review comprises of preparation and characterization of platelet membrane camouflaged nanoparticles and also focuses on their recent developments towards targeted therapy in cancer, immune diseases, atherosclerosis and phototherapy. Although platelet membrane camouflaged nanoparticles are currently in the preliminary stage of development, there is huge potential to explore this biodegradable and biocompatible delivery system.
Collapse
|
144
|
Wang J, Gu X, Ouyang Y, Chu L, Xu M, Wang K, Tong X. Engineering of Neutrophil Membrane Camouflaging Nanoparticles Realizes Targeted Drug Delivery for Amplified Antitumor Therapy. Int J Nanomedicine 2021; 16:1175-1187. [PMID: 33623381 PMCID: PMC7894798 DOI: 10.2147/ijn.s288636] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/23/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Although the neutrophil membrane (NM)-based nanoparticulate delivery system has exhibited rapid advances in tumor targeting stemmed from the inherited instinct, the antitumor effect requires further improvement due to inefficient cellular internalization in the absence of specific interactions between NM-coated nanoparticles and tumor cells. Methods Herein, we fabricated drug-paclitaxel loaded NM camouflaging nanoparticles (TNM-PN) modified with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), favorable for the cellular internalization. Results The results showed that TNM-PN exerted a significant cytotoxicity to tumor cells by TRAIL-mediated endocytosis and strong adhesion to inflamed endothelial cells in vitro. Due to TRAIL modification as well as the adhesive interactions between neutrophil and inflamed tumor vascular endothelial cells, tumors in TNM-PN group exhibited almost 2-fold higher fluorescence intensities than that of NM camouflaging nanoparticles and 3-fold higher than that of bare nanoparticles, respectively. Significant tumor inhibition and survival rates of mice were achieved in TNM-PN group as a consequence of prolonged blood circulations to 48 h and preferential tumor accumulations, which was ascribed to targeting adhesion originated from NM to immune evasion and subsequent excellent cellular internalization. Conclusion The research unveiled a novel strategy of amplifying cellular internalization based on NM coating nanotechnology to boost antitumor efficacy.
Collapse
Affiliation(s)
- Jingshuai Wang
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Xuemin Gu
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Yiqin Ouyang
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Lei Chu
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Mengjiao Xu
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Kun Wang
- Cancer Center, Shanghai East Hospital of Tongji University, Shanghai, People's Republic of China
| | - Xiaowen Tong
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
145
|
Chen F, Wang Y, Gao J, Saeed M, Li T, Wang W, Yu H. Nanobiomaterial-based vaccination immunotherapy of cancer. Biomaterials 2021; 270:120709. [PMID: 33581608 DOI: 10.1016/j.biomaterials.2021.120709] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapies including cancer vaccines, immune checkpoint blockade or chimeric antigen receptor T cells have been exploited as the attractive treatment modalities in recent years. Among these approaches, cancer vaccines that designed to deliver tumor antigens and adjuvants to activate the antigen presenting cells (APCs) and induce antitumor immune responses, have shown significant efficacy in inhibiting tumor growth, preventing tumor relapse and metastasis. Despite the potential of cancer vaccination strategies, the therapeutic outcomes in preclinical trials are failed to promote their clinical translation, which is in part due to their inefficient vaccination cascade of five critical steps: antigen identification, antigen encapsulation, antigen delivery, antigen release and antigen presentation to T cells. In recent years, it has been demonstrated that various nanobiomaterials hold great potential to enhance cancer vaccination cascade and improve their antitumor performance and reduce the off-target effect. We summarize the cutting-edge advances of nanobiomaterials-based vaccination immunotherapy of cancer in this review. The various cancer nanovaccines including antigen peptide/adjuvant-based nanovaccines, nucleic acid-based nanovaccines as well as biomimetic nanobiomaterials-based nanovaccines are discussed in detail. We also provide some challenges and perspectives associated with the clinical translation of cancer nanovaccines.
Collapse
Affiliation(s)
- Fangmin Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingjie Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Nano Science and Technology Institute, University of Science and Technology of China, Suzhou, 215123, China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Madiha Saeed
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tianliang Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
146
|
Zhang M, Cheng S, Jin Y, Zhang N, Wang Y. Membrane engineering of cell membrane biomimetic nanoparticles for nanoscale therapeutics. Clin Transl Med 2021; 11:e292. [PMID: 33635002 PMCID: PMC7819108 DOI: 10.1002/ctm2.292] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
In recent years, cell membrane camouflaging technology has emerged as an important strategy of nanomedicine, and the modification on the membranes is also a promising approach to enhance the properties of the nanoparticles, such as cancer targeting, immune evasion, and phototherapy sensitivity. Indeed, diversified approaches have been exploited to re-engineer the membranes of nanoparticles in several studies. In this review, first we discuss direct modification strategy of cell membrane camouflaged nanoparticles (CM-NP) via noncovalent, covalent, and enzyme-involved methods. Second, we explore how the membranes of CM-NPs can be re-engineered at the cellular level using strategies such as genetic engineering and membranes fusion. Due to the innate biological properties and excellent biocompatibility, the functionalized cell membrane-camouflaged nanoparticles have been widely applied in the fields of drug delivery, imaging, detoxification, detection, and photoactivatable therapy.
Collapse
Affiliation(s)
- Minghai Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yue Jin
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Nan Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yu Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
- Shanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| |
Collapse
|
147
|
Tang Y, Qian W, Zhang B, Liu W, Sun X, Ji W, Ma L, Zhu D. None-Loss Target Release of Biomimetic CaCO 3 Nanocomposites for Screening Bioactive Components and Target Proteins. ACS APPLIED BIO MATERIALS 2021. [DOI: 10.1021/acsabm.0c01197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yingying Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Wenhui Qian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
- Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, Jiangsu, China
| | - Bei Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Wenya Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
- Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, Jiangsu, China
| | - Xuetong Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Wenwen Ji
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Lijuan Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| |
Collapse
|
148
|
D'Souza JN, Prabhu A, Nagaraja GK, Navada K M, Kouser S, Manasa DJ. Unravelling the human triple negative breast cancer suppressive activity of biocompatible zinc oxide nanostructures influenced by Vateria indica (L.) fruit phytochemicals. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111887. [PMID: 33641895 DOI: 10.1016/j.msec.2021.111887] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/25/2020] [Accepted: 01/09/2021] [Indexed: 01/17/2023]
Abstract
The present study delineates the biosynthesis of ZnOVI nanostructures by using aqueous fruit extract of V. indica. The study has disclosed the role of V. indica fruit extract as both reducing and capping agents, ushering the formation of ZnOVI nanostructures with distinct morphologies. The formation of ZnOVI nanostructures was corroborated by FT-IR and UV-visible spectroscopy which was further substantiated by the elemental composition study through EDS spectroscopy. The nanostructures were also investigated by Rietveld refinement of PXRD data, FE-SEM, and BET analysis. The morphology, size, and surface area were found to be precursor stoichiometry dependent. The in-vitro cytotoxicity study of ZnOVI nanostructures carried out on MDA-MB468 human triple-negative breast cancer (TNBC) cells has revealed their potential cytotoxicity (91.18 ± 1.98). MTT assay performed on the NIH3T3 mouse fibroblast cells has unfolded the non-toxic nature of ZnOVI nanostructures. Additionally, the results of the AO-EB dual staining assay indicated early apoptosis in TNBC cells by displaying greenish yellow-fluorescence in the nuclei. Reactive oxygen species (ROS) measurement study has confirmed the elevated intracellular levels of ROS, supporting the oxidative-stress induced cytotoxicity in ZnOVI nanostructures treated TNBC cells. Furthermore, the haemocompatibility of ZnOVI nanostructures was evaluated using human erythrocytes. Thus, the obtained results have shown greater potential in the anticancer activity of bio-fabricated ZnOVI nanostructures.
Collapse
Affiliation(s)
- Josline Neetha D'Souza
- Department of Chemistry, Mangalore University, Mangaloagangothri 574199, Karnataka, India
| | - Ashwini Prabhu
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte 575018, Karnataka, India
| | - G K Nagaraja
- Department of Chemistry, Mangalore University, Mangaloagangothri 574199, Karnataka, India.
| | - Meghana Navada K
- Department of Chemistry, Mangalore University, Mangaloagangothri 574199, Karnataka, India
| | - Sabia Kouser
- Department of Chemistry, Mangalore University, Mangaloagangothri 574199, Karnataka, India
| | - D J Manasa
- Department of Botany, Davanagere University, Davanagere 577007, Karnataka, India
| |
Collapse
|
149
|
Liu W, Yan Q, Xia C, Wang X, Kumar A, Wang Y, Liu Y, Pan Y, Liu J. Recent advances in cell membrane coated metal-organic frameworks (MOFs) for tumor therapy. J Mater Chem B 2021; 9:4459-4474. [PMID: 33978055 DOI: 10.1039/d1tb00453k] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In improving the tumor-targeting ability of metal-organic frameworks (MOFs) for tumor therapy and avoiding the clearance as well as capture by the immune system, there are still several challenges, which limit the development and bio-applications of MOFs. To overcome these challenges, various targeted modification strategies have been proposed. Amongst all the strategies, a promising cell membrane coating method has been explored and utilized for the syntheses of new cell membrane biomimetic MOFs (CMMs). Through such coating, various source cell membranes (e.g., red blood cell, immune cell, cancer cell, platelet, and fusion cell membranes) can be endowed with excellent properties such as long blood circulation, immune escape, and targeting ability. In the presented perspective, the synthetic method, characterization, and research progress in tumor therapy based on CMMs have been summarized. This is because, like many other technologies, the cell membrane coating technology also has several challenges to overcome. Hence, addressing and overcoming such challenges will promote and extend the bio-applications of MOFs which in the future may become a prospective carrier for cancer nano-medicine. Finally, the prospects and challenges of utilizing CMMs for tumor therapy have been discussed.
Collapse
Affiliation(s)
- Weicong Liu
- Department of Pharmacy, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China.
| | - Qianwen Yan
- Department of Pathology, The First People's Hospital of Foshan, Foshan 528000, China
| | - Chen Xia
- Department of Pharmacy, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China.
| | - Xiaoxiong Wang
- School of Civil and Environmental Engineering, Shenzhen Polytechnic, Shenzhen 518055, China.
| | - Abhinav Kumar
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226 007, India
| | - Yan Wang
- Department of Pharmacy, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China.
| | - Yiwei Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Ying Pan
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Jianqiang Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
150
|
Xie Q, Liu Y, Long Y, Wang Z, Jiang S, Ahmed R, Daniyal M, Li B, Liu B, Wang W. Hybrid-cell membrane-coated nanocomplex-loaded chikusetsusaponin IVa methyl ester for a combinational therapy against breast cancer assisted by Ce6. Biomater Sci 2021; 9:2991-3004. [DOI: 10.1039/d0bm02211j] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hybrid-cell membrane coating nanocomplexes loading chikusetsusaponin IVa methyl ester for combinational therapy against breast cancer assisted with Ce6.
Collapse
|