1
|
Wang QH, Cheng S, Han CY, Yang S, Gao SR, Yin WZ, Song WZ. Tailoring cell-inspired biomaterials to fuel cancer therapy. Mater Today Bio 2025; 30:101381. [PMID: 39742146 PMCID: PMC11683242 DOI: 10.1016/j.mtbio.2024.101381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Cancer stands as a predominant cause of mortality across the globe. Traditional cancer treatments, including surgery, radiotherapy, and chemotherapy, are effective yet face challenges like normal tissue damage, complications, and drug resistance. Biomaterials, with their advantages of high efficacy, targeting, and spatiotemporal controllability, have been widely used in cancer treatment. However, the biocompatibility limitations of traditional synthetic materials have restricted their clinical translation and application. Natural cell-inspired biomaterials inherently possess the targeting abilities of cells, biocompatibility, and immune evasion capabilities. Therefore, cell-inspired biomaterials can be used alone or in combination with other drugs or treatment strategies for cancer therapy. In this review, we first introduce the timeline of key milestones in cell-inspired biomaterials for cancer therapy. Then, we describe the abnormalities in cancer including biophysics, cellular biology, and molecular biology aspects. Afterwards, we summarize the design strategies of cell-inspired antitumor biomaterials. Subsequently, we elaborate on the application of antitumor biomaterials inspired by various cell types. Finally, we explore the current challenges and prospects of cell-inspired antitumor materials. This review aims to provide new opportunities and references for the development of antitumor cell-inspired biomaterials.
Collapse
Affiliation(s)
- Qi-Hui Wang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shi Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Chun-Yu Han
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shuang Yang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Sheng-Rui Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wan-Zhong Yin
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wen-Zhi Song
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| |
Collapse
|
2
|
Feng L, Wang X, Gao Z, Tong Y, Yuan X, Wu T, Xia D, Hu Y. Enhancing Chemotherapy Efficacy via an Autologous Erythrocyte-Anchoring Strategy with a Closed-System Drug-Transfer Device. ACS Biomater Sci Eng 2025; 11:429-441. [PMID: 39696880 DOI: 10.1021/acsbiomaterials.4c02128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Chemotherapeutic drugs often fail to localize efficiently to tumors when administered intravenously, causing off-target effects. This study proposes an autologous erythrocyte (ER)-anchoring strategy to improve chemotherapy efficacy and reduce side effects. Utilizing a modified hemodialysis instrument, a closed-system drug-transfer device was developed for autologous ER procurement and immunogenicity mitigation. Doxorubicin (DOX) and indocyanine green (ICG) were encapsulated in autologous ERs and then modified with DSPE-PEG-FA. The final product, DOX-ICG@ER-D, was reintroduced into circulation to enhance chemotherapy. These obtained DOX-ICG@ER-D showed good stability, minimal cardiotoxicity, and extended circulation time. Compared to free DOX, DOX-ICG@ER-D had a higher accumulation of DOX in hepatocellular carcinoma and the release of DOX could be controlled by laser irradiation. Tumor-bearing rats treated by these DOX-ICG@ER-D demonstrated improved antitumor efficacy and reduced cardiotoxicity. Thus, this autologous ER-anchoring strategy offers a promising alternative to intravenous chemotherapy in the clinic.
Collapse
Affiliation(s)
- Lingzi Feng
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Xiangqian Wang
- Department of Radiotherapy, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong 226361, P. R. China
| | - Ziyi Gao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China
| | - Yuqing Tong
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
| | - Xiaopeng Yuan
- Department of Radiotherapy, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong 226361, P. R. China
| | - Ting Wu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China
| | - Donglin Xia
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Yong Hu
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
| |
Collapse
|
3
|
Meng X, Wang X, Zhang Z, Song L, Chen J. Recent Advancements of Nanomedicine in Breast Cancer Surgery. Int J Nanomedicine 2024; 19:14143-14169. [PMID: 39759962 PMCID: PMC11699852 DOI: 10.2147/ijn.s494364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
Breast cancer surgery plays a pivotal role in the multidisciplinary approaches. Surgical techniques and objectives are gradually shifting from tumor complete resection towards prolonging survival, improving cosmetic outcomes, and restoring the social and psychological well-being of patients. However, surgical treatment still faces challenges such as inadequate sensitivity in sentinel lymph node localization, the need to improve intraoperative tumor boundary localization imaging, postoperative scar healing, and the risk of recurrence, necessitating other adjunct measures for improvement. To address these challenges, specificity-optimized nanomedicines have been introduced into the surgical therapeutic landscape of breast cancer. In particular, this review involves starting with an overview of breast structure and the composition of the tumor microenvironment and then introducing the guiding principle and foundation for the design of nanomedicine. Moreover, we will take the order process of breast cancer surgery diagnosis and treatment as the starting point, and adaptively propose the roles and advantages of nanomedicine in addressing the corresponding issues. Furthermore, we also involved the prospects of utilizing advanced technological approaches. Overall, this review seeks to uncover the sophisticated design and strategies of nanomedicine from a clinical standpoint, address the challenges faced in surgical treatment, and provide insights into this subject matter.
Collapse
Affiliation(s)
- Xiangyue Meng
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xin Wang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zhihao Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Linlin Song
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jie Chen
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
4
|
Hadi Barhaghtalab R, Tanimowo Aiyelabegan H, Maleki H, Mirzavi F, Gholizadeh Navashenaq J, Abdi F, Ghaffari F, Vakili-Ghartavol R. Recent advances with erythrocytes as therapeutics carriers. Int J Pharm 2024; 665:124658. [PMID: 39236775 DOI: 10.1016/j.ijpharm.2024.124658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Erythrocytes have gained popularity as a natural option for in vivo drug delivery due to their advantages, which include lengthy circulation times, biocompatibility, and biodegradability. Consequently, the drug's pharmacokinetics and pharmacodynamics in red blood cells can be considerably up the dosage. Here, we provide an overview of the erythrocyte membrane's structure and discuss the characteristics of erythrocytes that influence their suitability as carrier systems. We also cover current developments in the erythrocyte-based nanocarrier, which could be used for both active and passive targeting of disease tissues, particularly those of the reticuloendothelial system (RES) and cancer tissues. We also go over the most recent discoveries about the in vivo and in vitro uses of erythrocytes for medicinal and diagnostic purposes. Moreover, the clinical relevance of erythrocytes is discussed in order to improve comprehension and enable the potential use of erythrocyte carriers in the management of various disorders.
Collapse
Affiliation(s)
| | | | - Hassan Maleki
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Fereshteh Abdi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Faezeh Ghaffari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roghayyeh Vakili-Ghartavol
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
6
|
Gao C, Liu Y, Zhang TL, Luo Y, Gao J, Chu JJ, Gong BF, Chen XH, Yin T, Zhang J, Yin Y. Biomembrane-Derived Nanoparticles in Alzheimer's Disease Therapy: A Comprehensive Review of Synthetic Lipid Nanoparticles and Natural Cell-Derived Vesicles. Int J Nanomedicine 2023; 18:7441-7468. [PMID: 38090364 PMCID: PMC10712251 DOI: 10.2147/ijn.s436774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Current therapies for Alzheimer's disease used in the clinic predominantly focus on reducing symptoms with limited capability to control disease progression; thus, novel drugs are urgently needed. While nanoparticles (liposomes, high-density lipoprotein-based nanoparticles) constructed with synthetic biomembranes have shown great potential in AD therapy due to their excellent biocompatibility, multifunctionality and ability to penetrate the BBB, nanoparticles derived from natural biomembranes (extracellular vesicles, cell membrane-based nanoparticles) display inherent biocompatibility, stability, homing ability and ability to penetrate the BBB, which may present a safer and more effective treatment for AD. In this paper, we reviewed the synthetic and natural biomembrane-derived nanoparticles that are used in AD therapy. The challenges associated with the clinical translation of biomembrane-derived nanoparticles and future perspectives are also discussed.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Yan Liu
- Department of Clinical Pharmacy, Shanghai Jiao Tong University of Medicine, Shanghai, People’s Republic of China
| | - Ting-Lin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Yi Luo
- Department of Clinical Pharmacy, Shanghai Jiao Tong University of Medicine, Shanghai, People’s Republic of China
- New Drug Discovery and Development, Biotheus Inc., Zhuhai, People’s Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Jian-Jian Chu
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Bao-Feng Gong
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Xiao-Han Chen
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Tong Yin
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| | - Jian Zhang
- Department of Clinical Pharmacy, Shanghai Jiao Tong University of Medicine, Shanghai, People’s Republic of China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
7
|
Huang Y, Nie X, Liu X, Liu Y, Yu H, Gao X. Development of a highly-efficient erythrocyte-drug covalent conjugation platform and its use in treating thrombotic disorders. Cell Res 2023; 33:887-890. [PMID: 37666976 PMCID: PMC10624666 DOI: 10.1038/s41422-023-00868-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/18/2023] [Indexed: 09/06/2023] Open
Affiliation(s)
- Yanjie Huang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiaoqian Nie
- School of Basic Medical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xuewen Liu
- School of Basic Medical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yuehua Liu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huifei Yu
- Westlake Therapeutics, Inc., Hangzhou, Zhejiang, China
| | - Xiaofei Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
De Castro F, Stefàno E, Fanizzi FP, Di Corato R, Abdalla P, Luchetti F, Nasoni MG, Rinaldi R, Magnani M, Benedetti M, Antonelli A. Compatibility of Nucleobases Containing Pt(II) Complexes with Red Blood Cells for Possible Drug Delivery Applications. Molecules 2023; 28:6760. [PMID: 37836603 PMCID: PMC10574024 DOI: 10.3390/molecules28196760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The therapeutic advantages of some platinum complexes as major anticancer chemotherapeutic agents and of nucleoside analogue-based compounds as essential antiviral/antitumor drugs are widely recognized. Red blood cells (RBCs) offer a potential new strategy for the targeted release of therapeutic agents due to their biocompatibility, which can protect loaded drugs from inactivation in the blood, thus improving biodistribution. In this study, we evaluated the feasibility of loading model nucleobase-containing Pt(II) complexes into human RBCs that were highly stabilized by four N-donors and susceptible to further modification for possible antitumor/antiviral applications. Specifically, platinum-based nucleoside derivatives [PtII(dien)(N7-Guo)]2+, [PtII(dien)(N7-dGuo)]2+, and [PtII(dien)(N7-dGTP)] (dien = diethylenetriamine; Guo = guanosine; dGuo = 2'-deoxy-guanosine; dGTP = 5'-(2'-deoxy)-guanosine-triphosphate) were investigated. These Pt(II) complexes were demonstrated to be stable species suitable for incorporation into RBCs. This result opens avenues for the possible incorporation of other metalated nucleobases analogues, with potential antitumor and/or antiviral activity, into RBCs.
Collapse
Affiliation(s)
- Federica De Castro
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Erika Stefàno
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Riccardo Di Corato
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia, 73010 Arnesano, Italy;
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
| | - Pasant Abdalla
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Francesca Luchetti
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Maria Gemma Nasoni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Rosaria Rinaldi
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
- Mathematics and Physics “E. De Giorgi” Department, University of Salento, Via Monteroni, 73100 Lecce, Italy
| | - Mauro Magnani
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Michele Benedetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Antonella Antonelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| |
Collapse
|
9
|
Feng C, Tan P, Nie G, Zhu M. Biomimetic and bioinspired nano-platforms for cancer vaccine development. EXPLORATION (BEIJING, CHINA) 2023; 3:20210263. [PMID: 37933383 PMCID: PMC10624393 DOI: 10.1002/exp.20210263] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2023]
Abstract
The advent of immunotherapy has revolutionized the treating modalities of cancer. Cancer vaccine, aiming to harness the host immune system to induce a tumor-specific killing effect, holds great promises for its broad patient coverage, high safety, and combination potentials. Despite promising, the clinical translation of cancer vaccines faces obstacles including the lack of potency, limited options of tumor antigens and adjuvants, and immunosuppressive tumor microenvironment. Biomimetic and bioinspired nanotechnology provides new impetus for the designing concepts of cancer vaccines. Through mimicking the stealth coating, pathogen recognition pattern, tissue tropism of pathogen, and other irreplaceable properties from nature, biomimetic and bioinspired cancer vaccines could gain functions such as longstanding, targeting, self-adjuvanting, and on-demand cargo release. The specific behavior and endogenous molecules of each type of living entity (cell or microorganism) offer unique features to cancer vaccines to address specific needs for immunotherapy. In this review, the strategies inspired by eukaryotic cells, bacteria, and viruses will be overviewed for advancing cancer vaccine development. Our insights into the future cancer vaccine development will be shared at the end for expediting the clinical translation.
Collapse
Affiliation(s)
- Chenchao Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Peng Tan
- Klarman Cell ObservatoryBroad Institute of MIT and HarvardCambridgeUSA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
- GBA Research Innovation Institute for NanotechnologyGuangzhouChina
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
| |
Collapse
|
10
|
Zhang C, Cha R, Wang C, Chen X, Li Z, Xie Q, Jia L, Sun Y, Hu Z, Zhang L, Zhou F, Zhang Y, Jiang X. Red blood cell membrane-functionalized Nanofibrous tubes for small-diameter vascular grafts. Biomaterials 2023; 297:122124. [PMID: 37087981 DOI: 10.1016/j.biomaterials.2023.122124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 03/23/2023] [Accepted: 04/08/2023] [Indexed: 04/25/2023]
Abstract
The off-the-shelf small-diameter vascular grafts (SDVGs) have inferior clinical efficacy. Red blood cell membrane (Rm) has easy availability and multiple bioactive components (such as phospholipids, proteins, and glycoproteins), which can improve the clinic's availability and patency of SDVGs. Here we developed a facile approach to preparing an Rm-functionalized poly-ε-caprolactone/poly-d-lysine (Rm@PCL/PDL) tube by co-incubation and single-step rolling. The integrity, stability, and bioactivity of Rm on Rm@PCL/PDL were evaluated. The revascularization of Rm@PCL/PDL tubes was studied by implantation in the carotid artery of rabbits. Rm@PCL/PDL can be quickly prepared and showed excellent bioactivity with good hemocompatibility and great anti-inflammatory. Rm@PCL/PDL tubes as the substitute for the carotid artery of rabbits had good patency and quick remodeling within 21 days. Rm, as a "self" biomaterial with high biosafety, provides a new and facile approach to developing personalized or universal SDVGs for the clinic, which is of great significance in cardiovascular regenerative medicine and organ chip.
Collapse
Affiliation(s)
- Chunliang Zhang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences (Beijing), No. 29 Xueyuan Road, Haidian District, Beijing, 100083, PR China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing, 100190, PR China
| | - Ruitao Cha
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing, 100190, PR China.
| | - Chunyuan Wang
- Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, PR China
| | - Xingming Chen
- PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang Beili, Chaoyang District, Beijing, 100101, PR China
| | - Zulan Li
- PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang Beili, Chaoyang District, Beijing, 100101, PR China
| | - Qian Xie
- Division of Nephrology, Peking University Third Hospital, No. 49 Huayuan Road North, Haidian District, Beijing, 100191, PR China
| | - Liujun Jia
- Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, PR China
| | - Yang Sun
- Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, PR China
| | - Zhan Hu
- Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, PR China
| | - Lin Zhang
- Department of Adult Cardiac Surgery, Faculty of Cardiovascular Disease, The Sixth Medical Center of the General Hospital of the People's Liberation Army of China, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China.
| | - Fengshan Zhou
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences (Beijing), No. 29 Xueyuan Road, Haidian District, Beijing, 100083, PR China.
| | - Yan Zhang
- Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, 100037, PR China.
| | - Xingyu Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong, 518055, PR China.
| |
Collapse
|
11
|
Li J, Wang D, Pottenburgh J, Bower AJ, Asanad S, Lai EW, Simon C, Im L, Huryn LA, Tao Y, Tam J, Saeedi OJ. Visualization of erythrocyte stasis in the living human eye in health and disease. iScience 2023; 26:105755. [PMID: 36594026 PMCID: PMC9803835 DOI: 10.1016/j.isci.2022.105755] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/25/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Blood cells trapped in stasis have been reported within the microcirculation, but their relevance to health and disease has not been established. In this study, we introduce an in vivo imaging approach that reveals the presence of a previously-unknown pool of erythrocytes in stasis, located within capillary segments of the CNS, and present in 100% of subjects imaged. These results provide a key insight that blood cells pause as they travel through the choroidal microvasculature, a vascular structure that boasts the highest blood flow of any tissue in the body. Demonstration of clinical utility using deep learning reveals that erythrocyte stasis is altered in glaucoma, indicating the possibility of more widespread changes in choroidal microvascular than previously realized. The ability to monitor the choroidal microvasculature at the single cell level may lead to novel strategies for tracking microvascular health in glaucoma, age-related macular degeneration, and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Joanne Li
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dongyi Wang
- Bioimaging and Machine Vision Laboratory, Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Jessica Pottenburgh
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew J. Bower
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Samuel Asanad
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric W. Lai
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Caroline Simon
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lily Im
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Laryssa A. Huryn
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yang Tao
- Bioimaging and Machine Vision Laboratory, Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Johnny Tam
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Osamah J. Saeedi
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Mayorova OA, Gusliakova OI, Prikhozhdenko ES, Verkhovskii RA, Bratashov DN. Magnetic Platelets as a Platform for Drug Delivery and Cell Trapping. Pharmaceutics 2023; 15:pharmaceutics15010214. [PMID: 36678843 PMCID: PMC9866132 DOI: 10.3390/pharmaceutics15010214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
The possibility of using magnetically labeled blood cells as carriers is a novel approach in targeted drug-delivery systems, potentially allowing for improved bloodstream delivery strategies. Blood cells already meet the requirements of biocompatibility, safety from clotting and blockage of small vessels. It would solve the important problem of the patient's immune response to embedded foreign carriers. The high efficiency of platelet loading makes them promising research objects for the development of personalized drug-delivery systems. We are developing a new approach to use platelets decorated with magnetic nanoparticles as a targeted drug-delivery system, with a focus on bloodstream delivery. Platelets are non-nuclear blood cells and are of great importance in the pathogenesis of blood-clotting disorders. In addition, platelets are able to attach to circulating tumor cells. In this article, we studied the effect of platelets labeled with BSA-modified magnetic nanoparticles on healthy and cancer cells. This opens up broad prospects for future research based on the delivery of specific active substances by this method.
Collapse
Affiliation(s)
- Oksana A. Mayorova
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia
- Department of General Educations, Saratov State Vavilov Agrarian University, 1 Theater Square, 410012 Saratov, Russia
- Correspondence: (O.A.M.); (D.N.B.)
| | - Olga I. Gusliakova
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia
| | | | - Roman A. Verkhovskii
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia
| | - Daniil N. Bratashov
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia
- Correspondence: (O.A.M.); (D.N.B.)
| |
Collapse
|
13
|
Ghasemzadeh T, Hasannia M, Abnous K, Taghdisi SM, Nekooei S, Nekooei N, Ramezani M, Alibolandi M. Preparation of targeted theranostic red blood cell membranes-based nanobubbles for treatment of colon adenocarcinoma. Expert Opin Drug Deliv 2023; 20:131-143. [PMID: 36427011 DOI: 10.1080/17425247.2022.2152792] [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: 11/27/2022]
Abstract
OBJECTIVES Designing and fabrication of theranostic systems based on nanoscale gaseous vesicular systems, named nanobubbles (NBs), attracted enormous interest in recent years. Biomimetic vesicular platform (V-RBC-M) can improve the pharmacokinetics of the prepared platform due to augmented circulation half-life, desirable biodegradability and biocompatibility and reduced immunogenicity. METHODS V-RBC-M were used for the encapsulation of lipophilic camptothecin (CPT) in the bilayer of vesicles through top-down method, followed by filling the core of V-RBC-M with inert SF6 gas to fabricate NBs with ultrasonic contrast enhancement capability (SF6-NB-CPT). In the next step, targeted NBs were formed via decoration of MUC1 aptamer on the surface of NBs (Apt-SF6-NB-CPT). RESULTS The designed bio-NBs indicated high encapsulation efficiency and the sustained release of CPT at pH 7.4. In vitro study demonstrated higher cellular uptake and cytotoxicity of Apt-SF6-NB-CPT compared to SF6-NB-CPT in MUC1-overexpressing cells (C26). In vivo antitumor efficacy of the prepared NBs on C26 bearing BALB/c mice showed greater therapeutic efficacy and survival rate for Apt-SF6-NB-CPT. In this regard, SF6-NB-CPT showed 58% tumor growth suppression while Apt-SF6-NB-CPT system provided 95% tumor growth suppression. Furthermore, echogenic capability of SF6-NB-CPT was demonstrated through in vitro and in vivo ultrasonic imaging. CONCLUSIONS Our finding demonstrated that the prepared targeted NBs are a promising theranostic platform with effective therapeutic and diagnotic potentials.
Collapse
Affiliation(s)
- Tahoora Ghasemzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maliheh Hasannia
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Alimardani V, Rahiminezhad Z, DehghanKhold M, Farahavar G, Jafari M, Abedi M, Moradi L, Niroumand U, Ashfaq M, Abolmaali SS, Yousefi G. Nanotechnology-based cell-mediated delivery systems for cancer therapy and diagnosis. Drug Deliv Transl Res 2023; 13:189-221. [PMID: 36074253 DOI: 10.1007/s13346-022-01211-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 12/13/2022]
Abstract
The global prevalence of cancer is increasing, necessitating new additions to traditional treatments and diagnoses to address shortcomings such as ineffectiveness, complications, and high cost. In this context, nano and microparticulate carriers stand out due to their unique properties such as controlled release, higher bioavailability, and lower toxicity. Despite their popularity, they face several challenges including rapid liver uptake, low chemical stability in blood circulation, immunogenicity concerns, and acute adverse effects. Cell-mediated delivery systems are important topics to research because of their biocompatibility, biodegradability, prolonged delivery, high loading capacity, and targeted drug delivery capabilities. To date, a variety of cells including blood, immune, cancer, and stem cells, sperm, and bacteria have been combined with nanoparticles to develop efficient targeted cancer delivery or diagnosis systems. The review paper aimed to provide an overview of the potential applications of cell-based delivery systems in cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Vahid Alimardani
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Rahiminezhad
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahvash DehghanKhold
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ghazal Farahavar
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Jafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Abedi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Moradi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Uranous Niroumand
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ashfaq
- University Centre for Research & Development (UCRD), Chandigarh University, Gharaun, Mohali, 140413, Punjab, India. .,Department of Biotechnology, Chandigarh University, Gharaun, Mohali, 140413, Punjab, India.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran. .,Center for Drug Delivery in Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Gholamhossein Yousefi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran. .,Center for Drug Delivery in Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
Meng D, Yang S, Yang Y, Zhang L, Cui L. Synergistic chemotherapy and phototherapy based on red blood cell biomimetic nanomaterials. J Control Release 2022; 352:146-162. [PMID: 36252749 DOI: 10.1016/j.jconrel.2022.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Novel drug delivery systems (DDSs) have become the mainstay of research in targeted cancer therapy. By combining different therapeutic strategies, potential DDSs and synergistic treatment approaches are needed to effectively deal with evolving drug resistance and the adverse effects of cancer. Nowadays, developing and optimizing human cell-based DDSs has become a new research strategy. Among them, red blood cells can be used as DDSs as they significantly enhance the pharmacokinetics of the transported drug cargo. Phototherapy, as a novel adjuvant in cancer treatment, can be divided into photodynamic therapy and photothermal therapy. Phototherapy using erythropoietic nanocarriers to mimic the unique properties of erythrocytes and overcome the limitations of existing DDSs shows excellent prospects in clinical settings. This review provides an overview of the development of photosensitizers and research on bio-nano-delivery systems based on erythrocytes and erythrocyte membranes that are used in achieving synergistic outcomes during phototherapy/chemotherapy.
Collapse
Affiliation(s)
- Di Meng
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China
| | - Shuoye Yang
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China.
| | - Yanan Yang
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China
| | - Lu Zhang
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China
| | - Lan Cui
- College of Bioengineering, Henan University of Technology, Zhengzhou, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou, PR China
| |
Collapse
|
16
|
Nazli A, He DL, Liao D, Khan MZI, Huang C, He Y. Strategies and progresses for enhancing targeted antibiotic delivery. Adv Drug Deliv Rev 2022; 189:114502. [PMID: 35998828 DOI: 10.1016/j.addr.2022.114502] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 01/24/2023]
Abstract
Antibiotic resistance is a global health issue and a potential risk for society. Antibiotics administered through conventional formulations are devoid of targeting effect and often spread to various undesired body sites, leading to sub-lethal concentrations at the site of action and thus resulting in emergence of resistance, as well as side effects. Moreover, we have a very slim antibiotic pipeline. Drug-delivery systems have been designed to control the rate, time, and site of drug release, and innovative approaches for antibiotic delivery provide a glint of hope for addressing these issues. This review elaborates different delivery strategies and approaches employed to overcome the limitations of conventional antibiotic therapy. These include antibiotic conjugates, prodrugs, and nanocarriers for local and targeted antibiotic release. In addition, a wide range of stimuli-responsive nanocarriers and biological carriers for targeted antibiotic delivery are discussed. The potential advantages and limitations of targeted antibiotic delivery strategies are described along with possible solutions to avoid these limitations. A number of antibiotics successfully delivered through these approaches with attained outcomes and potentials are reviewed.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China
| | - David L He
- College of Chemistry, University of California, Berkeley, CA 94720, United States
| | - Dandan Liao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China
| | | | - Chao Huang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China.
| |
Collapse
|
17
|
Vincy A, Mazumder S, Amrita, Banerjee I, Hwang KC, Vankayala R. Recent Progress in Red Blood Cells-Derived Particles as Novel Bioinspired Drug Delivery Systems: Challenges and Strategies for Clinical Translation. Front Chem 2022; 10:905256. [PMID: 35572105 PMCID: PMC9092017 DOI: 10.3389/fchem.2022.905256] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/08/2022] [Indexed: 11/24/2022] Open
Abstract
Red Blood Cells (RBCs)-derived particles are an emerging group of novel drug delivery systems. The natural attributes of RBCs make them potential candidates for use as a drug carrier or nanoparticle camouflaging material as they are innately biocompatible. RBCs have been studied for multiple decades in drug delivery applications but their evolution in the clinical arena are considerably slower. They have been garnering attention for the unique capability of conserving their membrane proteins post fabrication that help them to stay non-immunogenic in the biological environment prolonging their circulation time and improving therapeutic efficiency. In this review, we discuss about the synthesis, significance, and various biomedical applications of the above-mentioned classes of engineered RBCs. This article is focused on the current state of clinical translation and the analysis of the hindrances associated with the transition from lab to clinic applications.
Collapse
|
18
|
Li M, Xu X, Shi R, Li Y, Lin Q, Gong T, Sun X, Zhang Z, Zhang L. Smart erythrocyte-hitchhiking insulin delivery system for prolonged automatic blood glucose control. Biomater Sci 2022; 10:2638-2649. [PMID: 35416806 DOI: 10.1039/d2bm00280a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Long and automatic control of blood glucose levels in diabetic patients could solve the problems caused by frequent insulin injections. Herein, we exploited the protection potential of erythrocytes by a "hitchhiking" strategy to significantly prolong the blood circulation time of a specifically-designed smart hitchhiking insulin delivery system (SHIDS). In the SHIDS, insulin, glucose oxidase, and catalase were co-loaded into nanoparticles formed by modified chitosan. The free glucosamines in chitosan anchor glucose transporters on the surface of erythrocytes, allowing erythrocyte-hitchhiking in the blood flow. A high glucose level triggers quick insulin release from the SHIDS to reduce the glucose level, which then slows the insulin release. This closed-loop glucose regulation by the SHIDS effectively controlled blood glucose within the normal range for at least 24 h and under 250 mg dL-1 for ∼48 h with one injection. This injectable erythrocyte-hitchhiking nanoplatform, which achieves long-term and automatic blood glucose control, thus has potential for further development. As the carrier could be used for delivering other drugs/agents or interacting with other substances, the hitchhiking strategy is versatile and may be applied in other medical applications too.
Collapse
Affiliation(s)
- Min Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaomin Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Rongying Shi
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yuai Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Qing Lin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Ling Zhang
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| |
Collapse
|
19
|
Wang M, Zhao J, Jiang H, Wang X. Tumor-targeted nano-delivery system of therapeutic RNA. MATERIALS HORIZONS 2022; 9:1111-1140. [PMID: 35134106 DOI: 10.1039/d1mh01969d] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The birth of RNAi technology has pioneered actionability at the molecular level. Compared to DNA, RNA is less stable and therefore requires more demanding delivery vehicles. With their flexible size, shape, structure, and accessible surface modification, non-viral vectors show great promise for application in RNA delivery. Different non-viral vectors have different ways of binding to RNA. Low immunotoxicity gives RNA significant advantages in tumor treatment. However, the delivery of RNA still has many limitations in vivo. This manuscript summarizes the size-targeting dependence of different organs, followed by a summary of nanovesicles currently in or undergoing clinical trials. It also reviews all RNA delivery systems involved in the current study, including natural, bionic, organic, and inorganic systems. It summarizes the advantages and disadvantages of different delivery methods, which will be helpful for future RNA vehicle design. It is hoped that this will be helpful for gene therapy of clinical tumors.
Collapse
Affiliation(s)
- Maonan Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Jingzhou Zhao
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
20
|
Robert M, Laperrousaz B, Piedrahita D, Gautier EF, Nemkov T, Dupuy F, Nader E, Salnot V, Mayeux P, D'Alessandro A, Lavazec C, Joly P, Scheer A, Connes P, Cibiel A. Multiparametric characterization of red blood cell physiology after hypotonic dialysis based drug encapsulation process. Acta Pharm Sin B 2022; 12:2089-2102. [PMID: 35847505 PMCID: PMC9279626 DOI: 10.1016/j.apsb.2021.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 10/12/2021] [Indexed: 11/06/2022] Open
Abstract
Red blood cells (RBCs) can act as carriers for therapeutic agents and can substantially improve the safety, pharmacokinetics, and pharmacodynamics of many drugs. Maintaining RBCs integrity and lifespan is important for the efficacy of RBCs as drug carrier. We investigated the impact of drug encapsulation by hypotonic dialysis on RBCs physiology and integrity. Several parameters were compared between processed RBCs loaded with l-asparaginase ("eryaspase"), processed RBCs without drug and non-processed RBCs. Processed RBCs were less hydrated and displayed a reduction of intracellular content. We observed a change in the metabolomic but not in the proteomic profile of processed RBCs. Encapsulation process caused moderate morphological changes and was accompanied by an increase of RBCs-derived Extracellular Vesicles release. Despite a decrease in deformability, processed RBCs were not mechanically retained in a spleen-mimicking device and had increased surface-to-volume ratio and osmotic resistance. Processed RBCs half-life was not significantly affected in a mouse model and our previous phase 1 clinical study showed that encapsulation of asparaginase in RBCs prolonged its in vivo half-life compared to free forms. Our study demonstrated that encapsulation by hypotonic dialysis may affect certain characteristics of RBCs but does not significantly affect the in vivo longevity of RBCs or their drug carrier function.
Collapse
Affiliation(s)
- Mélanie Robert
- Erytech Pharma, Lyon 69008, France
- Laboratoire interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team « Vascular Biology and Red Blood Cell », Université Claude Bernard Lyon 1, Université de Lyon, Lyon 69008, France
- Labex GR-Ex, Paris 75015, France
| | | | | | - Emilie-Fleur Gautier
- Labex GR-Ex, Paris 75015, France
- 3P5 proteom'IC facility, Université de Paris, Institut Cochin, INSERM, CNRS, Paris 75014, France
| | - Travis Nemkov
- University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Omix Technologies, Inc., Aurora, CO 80045, USA
| | - Florian Dupuy
- Labex GR-Ex, Paris 75015, France
- Inserm U1016, CNRS 8104, Université de Paris, Institut Cochin, Paris 75014, France
| | - Elie Nader
- Laboratoire interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team « Vascular Biology and Red Blood Cell », Université Claude Bernard Lyon 1, Université de Lyon, Lyon 69008, France
- Labex GR-Ex, Paris 75015, France
| | - Virginie Salnot
- 3P5 proteom'IC facility, Université de Paris, Institut Cochin, INSERM, CNRS, Paris 75014, France
| | - Patrick Mayeux
- Labex GR-Ex, Paris 75015, France
- 3P5 proteom'IC facility, Université de Paris, Institut Cochin, INSERM, CNRS, Paris 75014, France
| | - Angelo D'Alessandro
- University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Omix Technologies, Inc., Aurora, CO 80045, USA
| | - Catherine Lavazec
- Labex GR-Ex, Paris 75015, France
- Inserm U1016, CNRS 8104, Université de Paris, Institut Cochin, Paris 75014, France
| | - Philippe Joly
- Laboratoire interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team « Vascular Biology and Red Blood Cell », Université Claude Bernard Lyon 1, Université de Lyon, Lyon 69008, France
- Labex GR-Ex, Paris 75015, France
- Laboratoire de Biochimie et de Biologie Moléculaire, UF de biochimie des pathologies érythrocytaires, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Bron 69500, France
| | | | - Philippe Connes
- Laboratoire interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team « Vascular Biology and Red Blood Cell », Université Claude Bernard Lyon 1, Université de Lyon, Lyon 69008, France
- Labex GR-Ex, Paris 75015, France
| | | |
Collapse
|
21
|
Yu Q, Li M, Chen H, Xu L, Cheng J, Lin G, Liu Y, Su Z, Yang X, Li Y, Chen J, Xie J. The discovery of berberine erythrocyte-hemoglobin self-assembly delivery system: a neglected carrier underlying its pharmacokinetics. Drug Deliv 2022; 29:856-870. [PMID: 35277093 PMCID: PMC8920379 DOI: 10.1080/10717544.2022.2036870] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Berberine (BBR) has extremely low concentration and high tissue distribution. However, current pharmacokinetic studies predominantly focus on its concentration in plasma, which could hardly make a comprehensive understanding of its pharmacokinetic process. This study made a pioneering endeavor to explore the erythrocyte-hemoglobin (Hb) self-assembly system of BBR by exploring the interaction of BBR with erythrocyte and the combination of BBR with Hb. Results showed that BBR had a low bioavailability (C0 = 2.833 μg/mL via intravenous administration of 2.5 mg/kg BBR and Cmax = 0.260 μg/mL via oral administration of 400 mg/kg BBR). Besides, BBR achieved higher concentrations in erythrocytes than plasma, and the erythrocytes count and Hb content were significantly decreased after intravenous administration. Hemolysis rate indicated the BBR-erythrocyte system (with 2% erythrocytes) was relatively stable without hemolysis at the concentration of 1.00 mg/mL. And the maximum percentage of drug loading was 100% when the BBR-erythrocyte concentration was 0.185 μg/mL. Furthermore, incubation of BBR and erythrocytes resulted in internalization of the erythrocyte membrane and the formation of intracellular vacuoles. The thermodynamic parameters indicated that the binding process of bovine hemoglobin (BHB) and BBR was spontaneous. UV-vis absorption spectra, synchronous fluorescence, circular dichroism and Raman spectra collectively indicated that BBR showed strong binding affinity toward BHB and affected the molecular environment of residues like tryptophan and tyrosine in BHB, resulting in the conformational changes of its secondary and tertiary structure. Molecular docking indicated BBR interacted with Arg-141 residue of BHB via hydrogen bond with the bond length of 2.55 Å. The ΔG value of the BHB-BBR system was −31.79 kJ/mol. Molecular dynamics simulation indicated the root mean square derivation of BBR-BHB was <0.025 nm, suggestive of stable conformation. Cumulatively, there was an erythrocyte-Hb self-assembled drug delivery system after oral or intravenous administration of BBR, which conceivably gained novel insight into the discrepancy between the extremely low plasma concentration and relatively high tissue concentration of BBR.
Collapse
Affiliation(s)
- Qiuxia Yu
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, China
| | - Minhua Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hanbin Chen
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lieqiang Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juanjuan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaobo Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|
22
|
Ci T, Zhang W, Qiao Y, Li H, Zang J, Li H, Feng N, Gu Z. Delivery strategies in treatments of leukemia. Chem Soc Rev 2022; 51:2121-2144. [PMID: 35188506 DOI: 10.1039/d1cs00755f] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leukemia is a hematological malignancy associated with the uncontrolled proliferation of mutant progenitors, suppressing the production of normal blood cells. Current treatments, including chemotherapy, radiotherapy, and immunotherapy, still lead to unsatisfactory results with a 5 year survival rate of only 30-50%. The poor prognosis is related to both disease relapse and treatment-associated toxicity. Delivery strategies can improve the in vivo pharmacokinetics of drugs, navigating the therapeutics to target cells or the tumor microenvironment and reversing drug resistance, which maximizes tumor elimination and alleviates systematic adverse effects. This review discusses available FDA-approved anti-leukemia drugs and therapies with a focus on the advances in the development of anti-leukemia drug delivery systems. Additionally, challenges in clinical translation of the delivery strategies and future research opportunities in leukemia treatment are also included.
Collapse
Affiliation(s)
- Tianyuan Ci
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wentao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yingyu Qiao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Jing Zang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hongjun Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China. .,Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
23
|
García-Álvarez R, Vallet-Regí M. Bacteria and cells as alternative nano-carriers for biomedical applications. Expert Opin Drug Deliv 2022; 19:103-118. [PMID: 35076351 PMCID: PMC8802895 DOI: 10.1080/17425247.2022.2029844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Nano-based systems have received a lot of attention owing to their particular properties and, hence, have been proposed for a wide variety of biomedical applications. These nanosystems could be potentially employed for diagnosis and therapy of different medical issues. Although these nanomaterials are designed for specific tasks, interactions, and transformations when administered to the human body affect their performance and behavior. In this regard, bacteria and other cells have been presented as alternative nanocarriers. These microorganisms can be genetically modified and customized for a more specific therapeutic action and, in combination with nanomaterials, can lead to bio-hybrids with a unique potential for biomedical purposes. AREAS COVERED Literature regarding bacteria and cells employed in combination with nanomaterials for biomedical applications is revised and discussed in this review. The potential as well as the limitations of these novel bio-hybrid systems are evaluated. Several examples are presented to show the performance of these alternative nanocarriers. EXPERT OPINION Bio-hybrid systems have shown their potential as alternative nanocarriers as they contribute to better performance than traditional nano-based systems. Nevertheless, their limitations must be studied, and advantages and drawbacks assessed before their application to medicine.
Collapse
Affiliation(s)
- Rafaela García-Álvarez
- Departamento de Química En Ciencias Farmacéuticas, Unidad de Química Inorgánica Y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre I+12, Madrid, Spain
- Ciber de Bioingeniería, Biomateriales Y Nanomedicina, Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química En Ciencias Farmacéuticas, Unidad de Química Inorgánica Y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre I+12, Madrid, Spain
- Ciber de Bioingeniería, Biomateriales Y Nanomedicina, Madrid, Spain
| |
Collapse
|
24
|
Resealed erythrocytes: Towards a novel approach for anticancer therapy. J INDIAN CHEM SOC 2021. [DOI: 10.1016/j.jics.2021.100257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
25
|
Erythrocyte-enabled immunomodulation for vaccine delivery. J Control Release 2021; 341:314-328. [PMID: 34838929 DOI: 10.1016/j.jconrel.2021.11.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022]
Abstract
Erythrocytes capture pathogens in circulation and present them to antigen-presenting cells (APCs) in the spleen. Senescent or apoptotic erythrocytes are physiologically eliminated by splenic APCs in a non-inflammatory manner as to not induce an immune reaction, while damaged erythrocytes tend to induce immune activation. The distinct characteristics of erythrocytes in their lifespan or different states inspire the design of targeting splenic APCs for vaccine delivery. Specifically, normal or damaged erythrocyte-driven immune targeting can induce antigen-specific immune activation, whereas senescent or apoptotic erythrocytes can be tailored to achieve antigen-specific immune tolerance. Recent studies have revealed the potential of erythrocyte-based vaccine delivery; however, there is still no in-depth review to describe the latest progress. This review summarizes the characteristics, different immune functions, and diverse vaccine delivery behaviors and biomedical applications of erythrocytes in different states. This review aims to contribute to the rational design and development of erythrocyte-based vaccine delivery systems for treating various infections, tumors, inflammatory diseases, and autoimmune diseases.
Collapse
|
26
|
Li Y, Raza F, Liu Y, Wei Y, Rong R, Zheng M, Yuan W, Su J, Qiu M, Li Y, Raza F, Liu Y, Wei Y, Rong R, Zheng M, Yuan W, Su J, Qiu M. Clinical progress and advanced research of red blood cells based drug delivery system. Biomaterials 2021; 279:121202. [PMID: 34749072 DOI: 10.1016/j.biomaterials.2021.121202] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 09/27/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023]
Abstract
Red blood cells (RBCs) are biocompatible carriers that can be employed to deliver different bioactive substances. In the past few decades, many strategies have been developed to encapsulate or attach drugs to RBCs. Osmotic-based encapsulation methods have been industrialized recently, and some encapsulated RBC formulations have reached the clinical stage for treating tumors and neurological diseases. Inspired by the intrinsic properties of intact RBCs, some advanced delivery strategies have also been proposed. These delivery systems combine RBCs with other novel systems to further exploit and expand the application of RBCs. This review summarizes the clinical progress of drugs encapsulated into intact RBCs, focusing on the loading and clinical trials. It also introduces the latest advanced research based on developing prospects and limitations of intact RBCs drug delivery system (DDS), hoping to provide a reference for related research fields and further application potential of intact RBCs based drug delivery system.
Collapse
Affiliation(s)
- Yichen Li
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Faisal Raza
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Yuhao Liu
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Yiqi Wei
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Ruonan Rong
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Mengyuan Zheng
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Weien Yuan
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Jing Su
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China.
| | - Mingfeng Qiu
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China.
| | - Y Li
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - F Raza
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Y Liu
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - Y Wei
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - R Rong
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - M Zheng
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - W Yuan
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - J Su
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| | - M Qiu
- School of Pharmacy Shanghai Jiao Tong University 800, Dongchuan Road, 200240, Shanghai, China
| |
Collapse
|
27
|
Chowdhury P, Ghosh U, Samanta K, Jaggi M, Chauhan SC, Yallapu MM. Bioactive nanotherapeutic trends to combat triple negative breast cancer. Bioact Mater 2021; 6:3269-3287. [PMID: 33778204 PMCID: PMC7970221 DOI: 10.1016/j.bioactmat.2021.02.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/27/2021] [Accepted: 02/28/2021] [Indexed: 02/09/2023] Open
Abstract
The management of aggressive breast cancer, particularly, triple negative breast cancer (TNBC) remains a formidable challenge, despite treatment advancement. Although newer therapies such as atezolizumab, olaparib, and sacituzumab can tackle the breast cancer prognosis and/or progression, but achieved limited survival benefit(s). The current research efforts are aimed to develop and implement strategies for improved bioavailability, targetability, reduce systemic toxicity, and enhance therapeutic outcome of FDA-approved treatment regimen. This review presents various nanoparticle technology mediated delivery of chemotherapeutic agent(s) for breast cancer treatment. This article also documents novel strategies to employ cellular and cell membrane cloaked (biomimetic) nanoparticles for effective clinical translation. These technologies offer a safe and active targeting nanomedicine for effective management of breast cancer, especially TNBC.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Upasana Ghosh
- Department of Biomedical Engineering, School of Engineering, Rutgers University, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Kamalika Samanta
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C. Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M. Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| |
Collapse
|
28
|
Surface loading of nanoparticles on engineered or natural erythrocytes for prolonged circulation time: strategies and applications. Acta Pharmacol Sin 2021; 42:1040-1054. [PMID: 33772141 DOI: 10.1038/s41401-020-00606-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/27/2020] [Indexed: 12/12/2022] Open
Abstract
Nano drug-delivery systems (DDS) may significantly improve efficiency and reduce toxicity of loaded drugs, but a few nano-DDS are highly successful in clinical use. Unprotected nanoparticles in blood flow are often quickly cleared, which could limit their circulation time and drug delivery efficiency. Elongating their blood circulation time may improve their delivery efficiency or grant them new therapeutic possibilities. Erythrocytes are abundant endogenous cells in blood and are continuously renewed, with a long life span of 100-120 days. Hence, loading nanoparticles on the surface of erythrocytes to protect the nanoparticles could be highly effective for enhancing their in vivo circulation time. One of the key questions here is how to properly attach nanoparticles on erythrocytes for different purposes and different types of nanoparticles to achieve ideal results. In this review, we describe various methods to attach nanoparticles and drugs to the erythrocyte surface, and discuss the key factors that influence the stability and circulation properties of the erythrocytes-based delivery system in vivo. These data show that using erythrocytes as a host for nanoparticles possesses great potential for further development.
Collapse
|
29
|
Della Pelle G, Kostevšek N. Nucleic Acid Delivery with Red-Blood-Cell-Based Carriers. Int J Mol Sci 2021; 22:5264. [PMID: 34067699 PMCID: PMC8156122 DOI: 10.3390/ijms22105264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has the potential to become a staple of 21st-century medicine. However, to overcome the limitations of existing gene-delivery therapies, that is, poor stability and inefficient and delivery and accumulation of nucleic acids (NAs), safe drug-delivery systems (DDSs) allowing the prolonged circulation and expression of the administered genes in vivo are needed. In this review article, the development of DDSs over the past 70 years is briefly described. Since synthetic DDSs can be recognized and eliminated as foreign substances by the immune system, new approaches must be found. Using the body's own cells as DDSs is a unique and exciting strategy and can be used in a completely new way to overcome the critical limitations of existing drug-delivery approaches. Among the different circulatory cells, red blood cells (RBCs) are the most abundant and thus can be isolated in sufficiently large quantities to decrease the complexity and cost of the treatment compared to other cell-based carriers. Therefore, in the second part, this article describes 70 years of research on the development of RBCs as DDSs, covering the most important RBC properties and loading methods. In the third part, it focuses on RBCs as the NA delivery system with advantages and drawbacks discussed to decide whether they are suitable for NA delivery in vivo.
Collapse
Affiliation(s)
- Giulia Della Pelle
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, Jamova Cesta 39, 1000 Ljubljana, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
| |
Collapse
|
30
|
Javed S, Alshehri S, Shoaib A, Ahsan W, Sultan MH, Alqahtani SS, Kazi M, Shakeel F. Chronicles of Nanoerythrosomes: An Erythrocyte-Based Biomimetic Smart Drug Delivery System as a Therapeutic and Diagnostic Tool in Cancer Therapy. Pharmaceutics 2021; 13:368. [PMID: 33802156 PMCID: PMC7998655 DOI: 10.3390/pharmaceutics13030368] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/29/2022] Open
Abstract
Recently, drug delivery using natural biological carriers has emerged as one of the most widely investigated topics of research. Erythrocytes, or red blood cells, can act as potential carriers for a wide variety of drugs, including anticancer, antibacterial, antiviral, and anti-inflammatory, along with various proteins, peptides, enzymes, and other macromolecules. The red blood cell-based nanocarrier systems, also called nanoerythrosomes, are nanovesicles poised with extraordinary features such as long blood circulation times, the ability to escape immune system, the ability to release the drug gradually, the protection of drugs from various endogenous factors, targeted and specified delivery of drugs, as well as possessing both therapeutic and diagnostic applications in various fields of biomedical sciences. Their journey over the last two decades is escalating with fast pace, ranging from in vivo to preclinical and clinical studies by encapsulating a number of drugs into these carriers. Being biomimetic nanoparticles, they have enhanced the stability profile of drugs and their excellent site-specific targeting ability makes them potential carrier systems in the diagnosis and therapy of wide variety of tumors including gliomas, lung cancers, breast cancers, colon cancers, gastric cancers, and other solid tumors. This review focuses on the most recent advancements in the field of nanoerythrosomes, as an excellent and promising nanoplatform for the novel drug delivery of various drugs particularly antineoplastic drugs along with their potential as a promising diagnostic tool for the identification of different tumors.
Collapse
Affiliation(s)
- Shamama Javed
- Department of Pharmaceutics, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (S.J.); (M.H.S.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.); (M.K.)
- Department of Pharmaceutical Sciences, College of Pharmacy, Almaarefa University, Riyadh 11597, Saudi Arabia
| | - Ambreen Shoaib
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (A.S.); (S.S.A.)
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia;
| | - Muhammad Hadi Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (S.J.); (M.H.S.)
| | - Saad Saeed Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; (A.S.); (S.S.A.)
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.); (M.K.)
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.); (M.K.)
| |
Collapse
|
31
|
|
32
|
Flegel WA, Srivastava K, Sissung TM, Goldspiel BR, Figg WD. Pharmacogenomics with red cells: a model to study protein variants of drug transporter genes. Vox Sang 2021; 116:141-154. [PMID: 32996603 PMCID: PMC9108996 DOI: 10.1111/vox.12999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/11/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022]
Abstract
The PharmacoScan pharmacogenomics platform screens for variation in genes that affect drug absorption, distribution, metabolism, elimination, immune adverse reactions and targets. Among the 1,191 genes tested on the platform, 12 genes are expressed in the red cell membrane: ABCC1, ABCC4, ABCC5, ABCG2, CFTR, SLC16A1, SLC19A1, SLC29A1, ATP7A, CYP4F3, EPHX1 and FLOT1. These genes represent 5 ATP-binding cassette proteins, 3 solute carrier proteins, 1 ATP transport protein and 3 genes associated with drug metabolism and adverse drug reactions. Only ABCG2 and SLC29A1 encode blood group systems, JR and AUG, respectively. We propose red cells as an ex vivo model system to study the effect of heritable variants in genes encoding the transport proteins on the pharmacokinetics of drugs. Altered pharmacodynamics in red cells could also cause adverse reactions, such as haemolysis, hitherto unexplained by other mechanisms.
Collapse
Affiliation(s)
- Willy Albert Flegel
- Department of Transfusion Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Kshitij Srivastava
- Department of Transfusion Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tristan Michael Sissung
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Barry Ronald Goldspiel
- Clinical Trials Operations and Informatics Branch, Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William Douglas Figg
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Cheng Z, Liu S, Wu X, Raza F, Li Y, Yuan W, Qiu M, Su J. Autologous erythrocytes delivery of berberine hydrochloride with long-acting effect for hypolipidemia treatment. Drug Deliv 2020; 27:283-291. [PMID: 32013620 PMCID: PMC7034074 DOI: 10.1080/10717544.2020.1716880] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Discovery of novel pharmacological effects of berberine hydrochloride (BH) has made its clinical application valuable. However, further development and applications of BH are hampered by its short half-life and the side effects associated with its intravenous (iv) injection. To improve the hypolipidemia efficacy and reduce side effects, we encapsulated BH into biocompatible red blood cells (RBCs) to explore its sustained-release effect by hypotonic pre-swelling method. From in vitro evaluation, BH loaded RBCs (BH-RBCs) presented similar morphology and osmotic fragility to native RBCs (NRBCs). After the loading process, the BH-RBCs maintained around 69% of Na+/K+-ATPase activity of NRBCs and phosphatidylserine externalization value of BH-RBCs was about 26.1 ± 2.9%. The survival test showed that the loaded cells could circulate in plasma for over 9 d. For in vivo evaluation, a series of tests including pharmacokinetics study and hypolipidemic effect were carried out to examine the long-acting effect of BH-RBCs. The results showed that the release of BH in the loaded cells could last for about 5 d and the hypolipidemic effect can still be observed on 5 d after injection. BH-loaded autologous erythrocytes seem to be a promising sustained releasing delivery system with long hypolipidemic effect.
Collapse
Affiliation(s)
- Zhongyao Cheng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Siyu Liu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yichen Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
34
|
Capossela S, Mathew V, Boos M, Bertolo A, Krupkova O, Stoyanov JV. Novel Fast and Reliable Method for Nano-Erythrosome Production Using Shear Force. Drug Des Devel Ther 2020; 14:4547-4560. [PMID: 33149552 PMCID: PMC7604965 DOI: 10.2147/dddt.s258368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/07/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose The production of nano-erythrosomes (NEs) by extrusion, which is considered the “gold standard”, has several disadvantages such as difficult equipment assembly, long procedure time, variable pressure, and problems with sterility. An alternative approach, using ultrasound probe, has been shown to overheat the sample and have suboptimal results compared to the extrusion method. In our study, we propose, develop, and test a new method for the fabrication of NEs based on shear force and then compare it to the “gold standard” extrusion approach. Methods The new method consists of mechanical shear force disruption of the hemoglobin-depleted erythrocyte ghost membranes, with the aid of a rotor stator based tissue homogenizer. Using the same batches of erythrocyte ghost membranes, we compared NEs produced by shear force to NEs produced by the well-established extrusion approach. NEs were characterized for yield, size, encapsulation efficiency, morphology, and stability by flow cytometry (FC), transmission electron microscopy (TEM), and zeta potential analysis. Results The shear force based process was easier to set up, significantly faster, had better sterility control, and decreased variability between batches. The shear force method generated NEs with the desired size distribution (particles diameter ~125 nm), which were morphologically and functionally equivalent to the NEs produced by extrusion. NEs produced by shear force were stable in terms of counts, size, and fluorescence intensity for 3 weeks at +4°C. Moreover, they showed colloidal stability and minimal influence to centrifugal stress, turbulence shock, and hemolytic potential. Conclusion The newly proposed shear force method allows faster, easier, and highly reproducible NEs production when compared to the conventional extrusion approach. The new setup allows simultaneous production of sterile batches of NEs, which have homogenous size distribution, good stability, and improved shelf life storage. The ability of the shear force method to process also high concentration samples indicates a future potential development of large-scale NEs production and industrial application, which has been a challenge for the extrusion method.
Collapse
Affiliation(s)
- Simona Capossela
- SCI Biobanking and Translational Medicine, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Vikas Mathew
- SCI Biobanking and Translational Medicine, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Manuela Boos
- Institute for Biomechanics, D-HEST, ETH Zurich, Zurich, Switzerland
| | - Alessandro Bertolo
- SCI Biobanking and Translational Medicine, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Olga Krupkova
- Institute for Biomechanics, D-HEST, ETH Zurich, Zurich, Switzerland.,Department of Spinal Surgery, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel & University Hospital Basel, Basel, Switzerland
| | - Jivko V Stoyanov
- SCI Biobanking and Translational Medicine, Swiss Paraplegic Research, Nottwil, Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM), Zurich, Switzerland
| |
Collapse
|
35
|
Hanley TM, Vankayala R, Mac JT, Lo DD, Anvari B. Acute Immune Response of Micro- and Nanosized Erythrocyte-Derived Optical Particles in Healthy Mice. Mol Pharm 2020; 17:3900-3914. [PMID: 32820927 PMCID: PMC9844151 DOI: 10.1021/acs.molpharmaceut.0c00641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Erythrocyte-derived particles activated by near-infrared (NIR) light present a platform for various phototheranostic applications. We have engineered such a platform with indocyanine green as the NIR-activated agent. A particular feature of these particles is that their diameters can be tuned from micro- to nanoscale, providing a potential capability for broad clinical utility ranging from vascular to cancer-related applications. An important issue related to clinical translation of these particles is their immunogenic effects. Herein, we have evaluated the early-induced innate immune response of these particles in healthy Swiss Webster mice following tail vein injection by measurements of specific cytokines in blood serum, the liver, and the spleen following euthanasia. In particular, we have investigated the effects of particle size and relative dose, time-dependent cytokine response for up to 6 h postinjection, functionalization of the nanosized particles with folate or Herceptin, and dual injections of the particles 1 week apart. Mean concentrations of interleukin (IL)-6, IL-10, tumor necrosis factor (TNF)-α, and monocyte chemoattractant protein (MCP)-1 in response to injection of microsized particles at the investigated relative doses were significantly lower than the corresponding mean concentrations induced by lipopolysaccharide (positive control) at 2 h. All investigated doses of the nanosized particles induced significantly higher concentrations of MCP-1 in the liver and the spleen as compared to phosphate buffer saline (PBS) (negative control) at 2 h. In response to micro- and nanosized particles at the highest investigated dose, there were significantly higher levels of TNF-α in blood serum at 2 and 6 h postinjection as compared to the levels associated with PBS treatment at these times. Whereas the mean concentration of TNF-α in the liver significantly increased between 2 and 6 h postinjection in response to the injection of the microsized particles, it was significantly reduced during this time interval in response to the injection of the nanosized particles. In general, functionalization of the nanosized particles was associated with a reduction of IL-6 and MCP-1 in blood serum, the liver, and the spleen, and TNF-α in blood serum. With the exception of IL-10 in the spleen in response to nanosized particles, the second injection of micro- or nanosized particles did not lead to significantly higher concentrations of other cytokines at the investigated dose as compared to a single injection.
Collapse
Affiliation(s)
- Taylor M. Hanley
- Department of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| | - Raviraj Vankayala
- Department of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| | - Jenny T. Mac
- Department of Biochemistry, University of California, Riverside, Riverside, California 92521, United States
| | - David D. Lo
- Department of Biomedical Sciences, University of California, Riverside, Riverside, California 92521, United States
| | - Bahman Anvari
- Department of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| |
Collapse
|
36
|
AlQahtani SA, Harisa GI, Alomrani AH, Alanazi FK, Badran MM. Improved pharmacokinetic and biodistribution of 5-fluorouracil loaded biomimetic nanoerythrocytes decorated nanocarriers for liver cancer treatment. Colloids Surf B Biointerfaces 2020; 197:111380. [PMID: 33068824 DOI: 10.1016/j.colsurfb.2020.111380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/31/2020] [Accepted: 09/26/2020] [Indexed: 01/06/2023]
Abstract
Nanoerythrocytes membrane (NEs) has recently been used to improve pharmacokinetics and biodistribution for successful drug therapy. NEs intended to enhance the drug targeting due to immune evasion and long circulation. In this work, NEs could serve as efficient 5- fluorouracil (5-FU) carriers to target liver cells. NEs decorated 5-FU-loaded chitosan coated-poly (lactide-co-glycolic acid) nanoparticles (5-FU-C-NPs-NEs), chitosomes (5-FU-C-LPs-NEs) and 5-FU-NEs were prepared by hypotonic lysis and extrusion procedures. Moreover, 5-FU loaded-chitosan coated 5-FU-NPs (5-FU-C-NPs) and chitosomes (5-FU-C-LPs) for the compared issues were prepared. They were characterized in terms of particle size, encapsulation efficiency (EE), membrane protein content, phosphatidylserine exposure, surface morphology, and in vitro release profiles. Also, their cytotoxic efficacy was determined. Furthermore, pharmacokinetics and biodistribution studies were investigated for optimized formulation. The results revealed that 5-FU-C-NPs-NEs have narrow particle size distribution, desirable EE%, and retained the erythrocyte membrane properties as confirmed by polyacrylamide gel electrophoresis (SDS-PAGE). Additionally, it displayed a sustained release profile up to 72 h of 5-FU-C-NPs-NEs compared to other formulations. In comparison to 5-FU solution and 5-FU-C-NPs, 5-FU-C-NPs-NEs extended the drug release time in vivo with highly uptake by the liver. These results suggest that the 5-FU-C-NPs-NEs could be used to deliver 5-FU and enhance its targetability to liver cancer.
Collapse
Affiliation(s)
- Saeed A AlQahtani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Saudi Food and Drug Authority, Drug Sector, P.O. Box 4904, Riyadh 13513, Saudi Arabia
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Department of Biochemistry, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Abdullah H Alomrani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Nanobiotechnology Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Fars K Alanazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohamed M Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Department of Pharmaceutics, College of Pharmacy, Al-Azhar University Cairo, Egypt.
| |
Collapse
|
37
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
38
|
Zargar SM, Hafshejani DK, Eskandarinia A, Rafienia M, Kharazi AZ. A Review of Controlled Drug Delivery Systems Based on Cells and Cell Membranes. JOURNAL OF MEDICAL SIGNALS & SENSORS 2019; 9:181-189. [PMID: 31544058 PMCID: PMC6743242 DOI: 10.4103/jmss.jmss_53_18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Novel drug delivery systems have ameliorated drugs’ pharmacokinetics and declined undesired ramifications while led to a better patient compliance by extending the time of release. In fact, although there has been a multitude of encouraging achievements in controlled drug release, the application of micro- and nano-carriers is confronted with some challenges such as rapid clearance and inefficient targeting. In addition, since cell systems can be an appropriate alternative to micro- and nano-particles, they have been used as biological carriers. In general, features such as stable release into blood, slow clearance, efficient targeting, and high biocompatibility are the main properties of cells applied as drug carriers. Furthermore, some cells such as erythrocytes, leukocytes, stem cells, and platelets have been used as release systems. Hence, most common cells that were used as aforementioned release systems are going to be presented in this review article.
Collapse
Affiliation(s)
- Seyed Mohammad Zargar
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran
| | - Darioush Khodabakhshi Hafshejani
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Asghar Eskandarinia
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohamad Rafienia
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anousheh Zargar Kharazi
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
39
|
Zhang F, Mundaca-Uribe R, Gong H, Esteban-Fernández de Ávila B, Beltrán-Gastélum M, Karshalev E, Nourhani A, Tong Y, Nguyen B, Gallot M, Zhang Y, Zhang L, Wang J. A Macrophage-Magnesium Hybrid Biomotor: Fabrication and Characterization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1901828. [PMID: 31070278 DOI: 10.1002/adma.201901828] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/15/2019] [Indexed: 05/26/2023]
Abstract
Magnesium (Mg)-based micromotors are combined with live macrophage (MΦ) cells to create a unique MΦ-Mg biohybrid motor system. The resulting biomotors possess rapid propulsion ability stemming from the Mg micromotors and the biological functions provided by the live MΦ cell. To prepare the biohybrid motors, Mg microparticles coated with titanium dioxide and poly(l-lysine) (PLL) layers are incubated with live MΦs at low temperature. The formation of such biohybrid motors depends on the relative size of the MΦs and Mg particles, with the MΦ swallowing up Mg particles smaller than 5 µm. The experimental results and numerical simulations demonstrate that the motion of MΦ-Mg motors is determined by the size of the Mg micromotor core and the position of the MΦ during the attachment process. The MΦ-Mg motors also perform biological functions related to free MΦs such as endotoxin neutralization. Cell membrane staining and toxin neutralization studies confirm that the MΦs maintain their viability and functionality (e.g., endotoxin neutralization) after binding to the Mg micromotors. This new MΦ-Mg motor design can be expanded to different types of living cells to fulfill diverse biological tasks.
Collapse
Affiliation(s)
- Fangyu Zhang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rodolfo Mundaca-Uribe
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Hua Gong
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Mara Beltrán-Gastélum
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Emil Karshalev
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Amir Nourhani
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Tong
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Bryan Nguyen
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mathieu Gallot
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yue Zhang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
40
|
Surface protein engineering increases the circulation time of a cell membrane-based nanotherapeutic. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:169-178. [DOI: 10.1016/j.nano.2019.02.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/31/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023]
|
41
|
Majumder J, Taratula O, Minko T. Nanocarrier-based systems for targeted and site specific therapeutic delivery. Adv Drug Deliv Rev 2019; 144:57-77. [PMID: 31400350 PMCID: PMC6748653 DOI: 10.1016/j.addr.2019.07.010] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/02/2019] [Accepted: 07/05/2019] [Indexed: 01/04/2023]
Abstract
Systemic drug delivery methods such as oral or parenteral administration of free drugs possess relatively low treatment efficiency and marked adverse side effects. The use of nanoparticles for drug delivery in most cases substantially enhances drug efficacy, improves pharmacokinetics and drug release and limits their side effects. However, further enhancement in drug efficacy and significant limitation of adverse side effects can be achieved by specific targeting of nanocarrier-based delivery systems especially in combination with local administration. The present review describes major advantages and limitations of organic and inorganic nanocarriers or living cell-based drug and nucleic acid delivery systems. Among these, different nanoparticles, supramolecular gels, therapeutic cells as living drug carriers etc. have emerged as a new frontier in modern medicine.
Collapse
Affiliation(s)
- Joydeb Majumder
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; Environmental and Occupational Health Science Institute, Piscataway, NJ 08854, USA.
| |
Collapse
|
42
|
Alshehab M, Nitin N. Encapsulation and release of curcumin using an intact milk fat globule delivery system. Food Funct 2019; 10:7121-7130. [DOI: 10.1039/c9fo00489k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Milk fat globule carriers were evaluated as an encapsulation system for curcumin. Partitioning is confirmed using fluorescence imaging. Release of curcumin under simulated gastrointestinal conditions and associated morphological changes to the carriers were evaluated.
Collapse
Affiliation(s)
- Maha Alshehab
- Department of Food Science and Technology
- University of California-Davis
- Davis
- USA
| | - Nitin Nitin
- Department of Food Science and Technology
- University of California-Davis
- Davis
- USA
- Department of Biological and Agricultural Engineering
| |
Collapse
|
43
|
Zhang X, Qiu M, Guo P, Lian Y, Xu E, Su J. Autologous Red Blood Cell Delivery of Betamethasone Phosphate Sodium for Long Anti-Inflammation. Pharmaceutics 2018; 10:pharmaceutics10040286. [PMID: 30567356 PMCID: PMC6320894 DOI: 10.3390/pharmaceutics10040286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 12/30/2022] Open
Abstract
Although glucocorticoids are highly effective in treating various types of inflammation such as skin disease, rheumatic disease, and allergic disease, their application have been seriously limited for their high incidence of side effects, particularly in long term treatment. To improve efficacy and reduce side effects, we encapsulated betamethasone phosphate (BSP) into biocompatible red blood cells (RBCs) and explored its long acting-effect. BSP was loaded into rat autologous erythrocytes by hypotonic preswelling method, and the loading amount was about 2.5 mg/mL cells. In vitro, BSP loaded RBCs (BSP-RBCs) presented similar morphology, osmotic fragility to native RBCs (NRBCs). After the loading process, the loaded cells can maintain around 70% of Na+/K+-ATPase activity of natural cells. In vivo, a series of tests including survival, pharmacokinetics, and anti-inflammatory effect were carried out to examine the long-acting effect of BSP-RBCs. The results shown that the loaded cells could circulate in plasma for over nine days, the release of BSP can last for over seven days and the anti-inflammatory effect can still be observed on day 5 after injection. Totally, BSP-loaded autologous erythrocytes seem to be a promising sustained releasing delivery system with long anti-inflammatory effect.
Collapse
Affiliation(s)
- Xiumei Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Pengcheng Guo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yumei Lian
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Enge Xu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
44
|
Lu Y, Hu Q, Jiang C, Gu Z. Platelet for drug delivery. Curr Opin Biotechnol 2018; 58:81-91. [PMID: 30529814 DOI: 10.1016/j.copbio.2018.11.010] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/14/2018] [Indexed: 01/09/2023]
Abstract
Platelets play a vital physiological role in hemostasis, inflammation and tissue regeneration, which are associated with wound healing as well as cancer development and metastasis. These years, a variety of platelet-mediated drug delivery approaches have been developed due to their unique properties, such as quick replenishment and site-specific activation/adhesion. In this Current Opinion, focuses are put on strategies leveraging the physiological functions of platelets for the design of drug delivery systems, including platelet engineering, platelet hitchhiking, membrane coating, synthetic platelet fabrication and platelet-triggered drug release for different applications.
Collapse
Affiliation(s)
- Yifei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Quanyin Hu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, United States
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, United States; California NanoSystems Institute, Jonsson Comprehensive Cancer Center, and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, United States.
| |
Collapse
|
45
|
Magnetically driven drug delivery systems improving targeted immunotherapy for colon-rectal cancer. J Control Release 2018; 280:76-86. [DOI: 10.1016/j.jconrel.2018.04.052] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 04/29/2018] [Indexed: 11/17/2022]
|
46
|
Charoenphol P, Oswalt K, Bishop CJ. Therapeutics incorporating blood constituents. Acta Biomater 2018; 73:64-80. [PMID: 29626699 DOI: 10.1016/j.actbio.2018.03.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/01/2018] [Accepted: 03/28/2018] [Indexed: 12/17/2022]
Abstract
Blood deficiency and dysfunctionality can result in adverse events, which can primarily be treated by transfusion of blood or the re-introduction of properly functioning sub-components. Blood constituents can be engineered on the sub-cellular (i.e., DNA recombinant technology) and cellular level (i.e., cellular hitchhiking for drug delivery) for supplementing and enhancing therapeutic efficacy, in addition to rectifying dysfunctioning mechanisms (i.e., clotting). Herein, we report the progress of blood-based therapeutics, with an emphasis on recent applications of blood transfusion, blood cell-based therapies and biomimetic carriers. Clinically translated technologies and commercial products of blood-based therapeutics are subsequently highlighted and perspectives on challenges and future prospects are discussed. STATEMENT OF SIGNIFICANCE Blood-based therapeutics is a burgeoning field and has advanced considerably in recent years. Blood and its constituents, with and without modification (i.e., combinatorial), have been utilized in a broad spectrum of pre-clinical and clinically-translated treatments. This review article summarizes the most up-to-date progress of blood-based therapeutics in the following contexts: synthetic blood substitutes, acellular/non-recombinant therapies, cell-based therapies, and therapeutic sub-components. The article subsequently discusses clinically-translated technologies and future prospects thereof.
Collapse
|
47
|
Shakya AK, Nandakumar KS. An update on smart biocatalysts for industrial and biomedical applications. J R Soc Interface 2018; 15:20180062. [PMID: 29491182 PMCID: PMC5832743 DOI: 10.1098/rsif.2018.0062] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/08/2018] [Indexed: 12/22/2022] Open
Abstract
Recently, smart biocatalysts, where enzymes are conjugated to stimuli-responsive (smart) polymers, have gained significant attention. Based on the presence or absence of external stimuli, the polymer attached to the enzyme changes its conformation to protect the enzyme from the external environment and regulate the enzyme activity, thus acting as a molecular switch. Owing to this behaviour, smart biocatalysts can be separated easily from a reaction mixture and re-used several times. Several such smart polymer-based biocatalysts have been developed for industrial and biomedical applications. In addition, they have been used in biosensors, biometrics and nano-electronic devices. This review article covers recent advances in developing different kinds of stimuli-responsive enzyme bioconjugates, including conjugation strategies, and their applications.
Collapse
Affiliation(s)
| | - Kutty Selva Nandakumar
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
- Department of Medical Biophysics and Biochemistry, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
48
|
Xu H, Medina-Sánchez M, Magdanz V, Schwarz L, Hebenstreit F, Schmidt OG. Sperm-Hybrid Micromotor for Targeted Drug Delivery. ACS NANO 2018; 12:327-337. [PMID: 29202221 DOI: 10.1021/acsnano.7b06398] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
A sperm-driven micromotor is presented as a targeted drug delivery system, which is appealing to potentially treat diseases in the female reproductive tract. This system is demonstrated to be an efficient drug delivery vehicle by first loading a motile sperm cell with an anticancer drug (doxorubicin hydrochloride), guiding it magnetically, to an in vitro cultured tumor spheroid, and finally freeing the sperm cell to deliver the drug locally. The sperm release mechanism is designed to liberate the sperm when the biohybrid micromotor hits the tumor walls, allowing it to swim into the tumor and deliver the drug through the sperm-cancer cell membrane fusion. In our experiments, the sperm cells exhibited a high drug encapsulation capability and drug carrying stability, conveniently minimizing toxic side effects and unwanted drug accumulation in healthy tissues. Overall, sperm cells are excellent candidates to operate in physiological environments, as they neither express pathogenic proteins nor proliferate to form undesirable colonies, unlike other cells or microorganisms. This sperm-hybrid micromotor is a biocompatible platform with potential application in gynecological healthcare, treating or detecting cancer or other diseases in the female reproductive system.
Collapse
Affiliation(s)
- Haifeng Xu
- Institute for Integrative Nanosciences, IFW Dresden , Helmholtzstraße 20, 01069 Dresden, Germany
| | - Mariana Medina-Sánchez
- Institute for Integrative Nanosciences, IFW Dresden , Helmholtzstraße 20, 01069 Dresden, Germany
| | - Veronika Magdanz
- Institute for Integrative Nanosciences, IFW Dresden , Helmholtzstraße 20, 01069 Dresden, Germany
| | - Lukas Schwarz
- Institute for Integrative Nanosciences, IFW Dresden , Helmholtzstraße 20, 01069 Dresden, Germany
| | - Franziska Hebenstreit
- Institute for Integrative Nanosciences, IFW Dresden , Helmholtzstraße 20, 01069 Dresden, Germany
| | - Oliver G Schmidt
- Institute for Integrative Nanosciences, IFW Dresden , Helmholtzstraße 20, 01069 Dresden, Germany
- Material Systems for Nanoelectronics, Chemnitz University of Technology , Reichenhainer Straße 70, 09107 Chemnitz, Germany
| |
Collapse
|
49
|
Affiliation(s)
- Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
50
|
Macromolecular Conjugate and Biological Carrier Approaches for the Targeted Delivery of Antibiotics. Antibiotics (Basel) 2017; 6:antibiotics6030014. [PMID: 28677631 PMCID: PMC5617978 DOI: 10.3390/antibiotics6030014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/24/2017] [Accepted: 06/29/2017] [Indexed: 01/21/2023] Open
Abstract
For the past few decades, the rapid rise of antibiotic multidrug-resistance has presented a palpable threat to human health worldwide. Meanwhile, the number of novel antibiotics released to the market has been steadily declining. Therefore, it is imperative that we utilize innovative approaches for the development of antimicrobial therapies. This article will explore alternative strategies, namely drug conjugates and biological carriers for the targeted delivery of antibiotics, which are often eclipsed by their nanomedicine-based counterparts. A variety of macromolecules have been investigated as conjugate carriers, but only those most widely studied in the field of infectious diseases (e.g., proteins, peptides, antibodies) will be discussed in detail. For the latter group, blood cells, especially erythrocytes, have been successfully tested as homing carriers of antimicrobial agents. Bacteriophages have also been studied as a candidate for similar functions. Once these alternative strategies receive the amount of research interest and resources that would more accurately reflect their latent applicability, they will inevitably prove valuable in the perennial fight against antibiotic resistance.
Collapse
|