1
|
Xu C, Chen L, Liu G, Xu J, Lv W, Gao X, Xu P, Tang M, Wang Y, Zhao X, Nie G, Cheng K, Liu F. Tailoring an intravenously injectable oncolytic virus for augmenting radiotherapy. Cell Rep Med 2025:102078. [PMID: 40233744 DOI: 10.1016/j.xcrm.2025.102078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/19/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
Oncolytic viruses (OVs) combined with radiotherapy (RT) show promise but are limited by challenges such as poor intravenous delivery and insufficient RT-induced DNA damage. In this study, an oncolytic adenovirus (AD) formulation, RadioOnco (AD@PSSP), is developed to improve delivery, infectivity, immune response, and RT efficacy. The multifunctional polyethylenimine (PEI)-selenium-polyethylene glycol (PEG) (PSSP) enhances intravenous delivery, shields the virus from rapid clearance, and enables targeted delivery to tumor sites after RT. The exposed PEI enhances the infectivity of AD through electrostatic interactions, thereby increasing DNA damage after RT by inhibiting the expression of DNA repair proteins, such as CHEK1 and CDK1. Furthermore, AD-PEI captures and delivers RT-induced tumor-released antigens to lymph nodes, activating robust anti-tumor immune responses. Animal model data demonstrate that RadioOnco overcomes RT resistance, targets distant metastases, and promotes long-term immunity, addressing metastasis and recurrence. In summary, this intravenously injectable OV enhances RT synergy through surface modification with multifunctional materials.
Collapse
Affiliation(s)
- Chen Xu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Liting Chen
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Peijun Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ming Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yaohe Wang
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China.
| |
Collapse
|
2
|
Liu S, Li H, Xi S, Zhang Y, Sun T. Advancing CNS Therapeutics: Enhancing Neurological Disorders with Nanoparticle-Based Gene and Enzyme Replacement Therapies. Int J Nanomedicine 2025; 20:1443-1490. [PMID: 39925682 PMCID: PMC11806685 DOI: 10.2147/ijn.s457393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 12/12/2024] [Indexed: 02/11/2025] Open
Abstract
Given the complexity of the central nervous system (CNS) and the diversity of neurological conditions, the increasing prevalence of neurological disorders poses a significant challenge to modern medicine. These disorders, ranging from neurodegenerative diseases to psychiatric conditions, not only impact individuals but also place a substantial burden on healthcare systems and society. A major obstacle in treating these conditions is the blood-brain barrier (BBB), which restricts the passage of therapeutic agents to the brain. Nanotechnology, particularly the use of nanoparticles (NPs), offers a promising solution to this challenge. NPs possess unique properties such as small size, large surface area, and modifiable surface characteristics, enabling them to cross the BBB and deliver drugs directly to the affected brain regions. This review focuses on the application of NPs in gene therapy and enzyme replacement therapy (ERT) for neurological disorders. Gene therapy involves altering or manipulating gene expression and can be enhanced by NPs designed to carry various genetic materials. Similarly, NPs can improve the efficacy of ERT for lysosomal storage disorders (LSDs) by facilitating enzyme delivery to the brain, overcoming issues like immunogenicity and instability. Taken together, this review explores the potential of NPs in revolutionizing treatment options for neurological disorders, highlighting their advantages and the future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Haisong Li
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Shiwen Xi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- International Center of Future Science, Jilin University, Changchun, People’s Republic of China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
3
|
Xu H, You R, Zhang H, Wei W, Li T, Duan X. One-step on-chip preparation of nanoparticle-conjugated red blood cell carriers. Colloids Surf B Biointerfaces 2025; 246:114373. [PMID: 39556898 DOI: 10.1016/j.colsurfb.2024.114373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/02/2024] [Accepted: 11/09/2024] [Indexed: 11/20/2024]
Abstract
Red blood cell (RBC)-based carriers have emerged as promising vehicles for drug delivery due to their inherent biocompatibility and biodegradability. Traditional methods for loading nanoparticles (NPs) onto RBC surfaces often involve labor-intensive processes like incubation and multiple centrifugation steps, limiting their practicality and controllability. In this study, we introduce a fully integrated acoustofluidic platform that enables one-step preparation of NP-loaded RBC carriers with controlled modification and on-site purification. By incorporating a high-frequency bulk acoustic wave (BAW) resonator into a microfluidic chip, we utilize acoustic streaming effects to manipulate the movement and interaction of RBCs and NPs within the microchannel. This design allows for precise control over NP loading efficiency by adjusting the input power to the resonator. Experimental results using 200 nm positively charged fluorescent NPs demonstrate that our platform significantly enhances the interaction between RBCs and NPs, achieving efficient and controllable surface loading of NPs onto RBCs. Furthermore, the platform simplifies post-processing by directing excess NPs to waste outlets, eliminating the need for repetitive washing and centrifugation. This acoustofluidics approach not only automates the loading process but also offers high controllability, highlighting its potential for various applications in particle and cell surface modification.
Collapse
Affiliation(s)
- Huihui Xu
- State Key Laboratory of Precision Measuring Technology & Instruments, and College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Rui You
- State Key Laboratory of Precision Measuring Technology & Instruments, and College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Huijing Zhang
- State Key Laboratory of Precision Measuring Technology & Instruments, and College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Wei Wei
- State Key Laboratory of Precision Measuring Technology & Instruments, and College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Tiechuan Li
- State Key Laboratory of Precision Measuring Technology & Instruments, and College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| | - Xuexin Duan
- State Key Laboratory of Precision Measuring Technology & Instruments, and College of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
4
|
Zhang S, Fang H, Tian H. Recent Advances in Degradable Biomedical Polymers for Prevention, Diagnosis and Treatment of Diseases. Biomacromolecules 2024; 25:7015-7057. [PMID: 39420482 DOI: 10.1021/acs.biomac.4c01193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Biomedical polymers play a key role in preventing, diagnosing, and treating diseases, showcasing a wide range of applications. Their unique advantages, such as rich source, good biocompatibility, and excellent modifiability, make them ideal biomaterials for drug delivery, biomedical imaging, and tissue engineering. However, conventional biomedical polymers suffer from poor degradation in vivo, increasing the risks of bioaccumulation and potential toxicity. To address these issues, degradable biomedical polymers can serve as an alternative strategy in biomedicine. Degradable biomedical polymers can efficiently relieve bioaccumulation in vivo and effectively reduce patient burden in disease management. This review comprehensively introduces the classification and properties of biomedical polymers and the recent research progress of degradable biomedical polymers in various diseases. Through an in-depth analysis of their classification, properties, and applications, we aim to provide strong guidance for promoting basic research and clinical translation of degradable biomedical polymers.
Collapse
Affiliation(s)
- Siting Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
5
|
Cheng R, Wang S. Cell-mediated nanoparticle delivery systems: towards precision nanomedicine. Drug Deliv Transl Res 2024; 14:3032-3054. [PMID: 38615157 PMCID: PMC11445310 DOI: 10.1007/s13346-024-01591-0] [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] [Accepted: 03/24/2024] [Indexed: 04/15/2024]
Abstract
Cell-mediated nanoparticle delivery systems (CMNDDs) utilize cells as carriers to deliver the drug-loaded nanoparticles. Unlike the traditional nanoparticle drug delivery approaches, CMNDDs take the advantages of cell characteristics, such as the homing capabilities of stem cells, inflammatory chemotaxis of neutrophils, prolonged blood circulation of red blood cells, and internalization of macrophages. Subsequently, CMNDDs can easily prolong the blood circulation, cross biological barriers, such as the blood-brain barrier and the bone marrow-blood barrier, and rapidly arrive at the diseased areas. Such advantageous properties make CMNDDs promising delivery candidates for precision targeting. In this review, we summarize the recent advances in CMNDDs fabrication and biomedical applications. Specifically, ligand-receptor interactions, non-covalent interactions, covalent interactions, and internalization are commonly applied in constructing CMNDDs in vitro. By hitchhiking cells, such as macrophages, red blood cells, monocytes, neutrophils, and platelets, nanoparticles can be internalized or attached to cells to construct CMNDDs in vivo. Then we highlight the recent application of CMNDDs in treating different diseases, such as cancer, central nervous system disorders, lung diseases, and cardiovascular diseases, with a brief discussion about challenges and future perspectives in the end.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland.
| |
Collapse
|
6
|
Bezze A, Mattioda C, Ciardelli G, Mattu C. Harnessing cells to improve transport of nanomedicines. Eur J Pharm Biopharm 2024; 203:114446. [PMID: 39122052 DOI: 10.1016/j.ejpb.2024.114446] [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: 06/07/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Efficient tumour treatment is hampered by the poor selectivity of anticancer drugs, resulting in scarce tumour accumulation and undesired off-target effects. Nano-sized drug-delivery systems in the form of nanoparticles (NPs) have been proposed to improve drug distribution to solid tumours, by virtue of their ability of passive and active tumour targeting. Despite these advantages, literature studies indicated that less than 1% of the administered NPs can successfully reach the tumour mass, highlighting the necessity for more efficient drug transporters in cancer treatment. Living cells, such as blood cells, circulating immune cells, platelets, and stem cells, are often found as an infiltrating component in most solid tumours, because of their ability to naturally circumvent immune recognition, bypass biological barriers, and reach inaccessible tissues through innate tropism and active motility. Therefore, the tumour-homing ability of these cells can be harnessed to design living cell carriers able to improve the transport of drugs and NPs to tumours. Albeit promising, this approach is still in its beginnings and suffers from difficult scalability, high cost, and poor reproducibility. In this review, we present an overview of the most common cell transporters of drugs and NPs, and we discuss how different cell types interact with biological barriers to deliver cargoes of various natures to tumours. Finally, we analyse the different techniques used to load drugs or NPs in living cells and discuss their advantages and disadvantages.
Collapse
Affiliation(s)
- Andrea Bezze
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Carlotta Mattioda
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Gianluca Ciardelli
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Clara Mattu
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
7
|
Duan M, Cao R, Yang Y, Chen X, Liu L, Ren B, Wang L, Goh BC. Blood-Brain Barrier Conquest in Glioblastoma Nanomedicine: Strategies, Clinical Advances, and Emerging Challenges. Cancers (Basel) 2024; 16:3300. [PMID: 39409919 PMCID: PMC11475686 DOI: 10.3390/cancers16193300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is a prevalent type of malignancy within the central nervous system (CNS) that is associated with a poor prognosis. The standard treatment for GBM includes the surgical resection of the tumor, followed by radiotherapy and chemotherapy; yet, despite these interventions, overall treatment outcomes remain suboptimal. The blood-brain barrier (BBB), which plays a crucial role in maintaining the stability of brain tissue under normal physiological conditions of the CNS, also poses a significant obstacle to the effective delivery of therapeutic agents to GBMs. Recent preclinical studies have demonstrated that nanomedicine delivery systems (NDDSs) offer promising results, demonstrating both effective GBM targeting and safety, thereby presenting a potential solution for targeted drug delivery. In this review, we first explore the various strategies employed in preclinical studies to overcome the BBB for drug delivery. Subsequently, the results of the clinical translation of NDDSs are summarized, highlighting the progress made. Finally, we discuss potential strategies for advancing the development of NDDSs and accelerating their translational research through well-designed clinical trials in GBM therapy.
Collapse
Affiliation(s)
- Mengyun Duan
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Ruina Cao
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China;
| | - Yuan Yang
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Xiaoguang Chen
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lian Liu
- Department of Pharmacology, Health Science Center, Yangtze University, Jingzhou 434023, China;
| | - Boxu Ren
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - 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, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Boon-Cher Goh
- 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, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
8
|
Sadri M, Heidari S, Faridzadeh A, Roozbehani M, Toosi S, Mahmoudian RA, Hoseinzadeh A, Salmani Fard MT, Arab FL, Fard SR, Faraji F. Potential applications of macrophages in cancer immunotherapy. Biomed Pharmacother 2024; 178:117161. [PMID: 39047419 DOI: 10.1016/j.biopha.2024.117161] [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: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
Immunotherapy has improved cancer treatment based on investigations of tumor immune escape. Manipulation of the immune system stimulates antitumor immune responses and blocks tumor immune escape routes. Genetically adoptive cell therapy, such as T cells, has yielded promising results for hematologic malignancies, but their application to solid tumors has been challenging. Macrophages have a wide broad of capabilities in regulating immune responses, homeostasis, and tissue development, as well as the ability to phagocyte, present antigens, and infiltrate the tumor microenvironment (TME). Given the importance of macrophages in cancer development, they could serve as novel tool for tumor treatment. Therefore, macrophages are used in different formats for direct and indirect targeting of tumor cells. This review summarized the available data on the various applications of macrophages in cancer immunotherapy.
Collapse
Affiliation(s)
- Maryam Sadri
- Department of Immunology, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 1449614535, Tehran, Iran.
| | - Sahel Heidari
- Department of Immunology, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 1449614535, Tehran, Iran.
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad 1313199137, Iran.
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 1449614535, Tehran, Iran.
| | - Shirin Toosi
- Stem Cell and Regenerative Medicine Center, Mashhad University of Medical Science, Mashhad 1313199137, Iran.
| | | | - Akram Hoseinzadeh
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 3513119111, Iran.
| | - Mohammad Taha Salmani Fard
- School of Biology, College of Science, University of Tehran, Faculty of Sciences, Enqelab Square, Tehran 1417614411, Iran.
| | - Fahimeh Lavi Arab
- Immunology Research center, Mashhad University of Medical Sciences, Mashhad 1313199137, Iran.
| | - Soheil Rahmani Fard
- Antimicrobial Resistance Research Center, Institute of Immunology and Infection Diseases Iran University of Medical Sciences, Floor 3, Building no. 3, Hazrat-e Rasool General Hospital, Niyayesh St, Sattar Khan St, P.O. Box: 1445613131, Tehran, Iran.
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infection Diseases Iran University of Medical Sciences, Floor 3, Building no. 3, Hazrat-e Rasool General Hospital, Niyayesh St, Sattar Khan St, P.O. Box: 1445613131, Tehran, Iran.
| |
Collapse
|
9
|
Peng X, Fang J, Lou C, Yang L, Shan S, Wang Z, Chen Y, Li H, Li X. Engineered nanoparticles for precise targeted drug delivery and enhanced therapeutic efficacy in cancer immunotherapy. Acta Pharm Sin B 2024; 14:3432-3456. [PMID: 39220871 PMCID: PMC11365410 DOI: 10.1016/j.apsb.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 09/04/2024] Open
Abstract
The advent of cancer immunotherapy has imparted a transformative impact on cancer treatment paradigms by harnessing the power of the immune system. However, the challenge of practical and precise targeting of malignant cells persists. To address this, engineered nanoparticles (NPs) have emerged as a promising solution for enhancing targeted drug delivery in immunotherapeutic interventions, owing to their small size, low immunogenicity, and ease of surface modification. This comprehensive review delves into contemporary research at the nexus of NP engineering and immunotherapy, encompassing an extensive spectrum of NP morphologies and strategies tailored toward optimizing tumor targeting and augmenting therapeutic effectiveness. Moreover, it underscores the mechanisms that NPs leverage to bypass the numerous obstacles encountered in immunotherapeutic regimens and probes into the combined potential of NPs when co-administered with both established and novel immunotherapeutic modalities. Finally, the review evaluates the existing limitations of NPs as drug delivery platforms in immunotherapy, which could shape the path for future advancements in this promising field.
Collapse
Affiliation(s)
- Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Jianjun Fang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Chuyuan Lou
- Department of Ophthalmology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710004, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Shaobo Shan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 10050, China
| | - Zixian Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, China
| | - Yutong Chen
- Department of Pathology, Medical College, Jinan University, Guangzhou 510632, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm SE-17177, Sweden
| |
Collapse
|
10
|
Guo Q, Qian ZM. Macrophage based drug delivery: Key challenges and strategies. Bioact Mater 2024; 38:55-72. [PMID: 38699242 PMCID: PMC11061709 DOI: 10.1016/j.bioactmat.2024.04.004] [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: 12/11/2023] [Revised: 03/14/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
As a natural immune cell and antigen presenting cell, macrophages have been studied and engineered to treat human diseases. Macrophages are well-suited for use as drug carriers because of their biological characteristics, such as excellent biocompatibility, long circulation, intrinsic inflammatory homing and phagocytosis. Meanwhile, macrophages' uniquely high plasticity and easy re-education polarization facilitates their use as part of efficacious therapeutics for the treatment of inflammatory diseases or tumors. Although recent studies have demonstrated promising advances in macrophage-based drug delivery, several challenges currently hinder further improvement of therapeutic effect and clinical application. This article focuses on the main challenges of utilizing macrophage-based drug delivery, from the selection of macrophage sources, drug loading, and maintenance of macrophage phenotypes, to drug migration and release at target sites. In addition, corresponding strategies and insights related to these challenges are described. Finally, we also provide perspective on shortcomings on the road to clinical translation and production.
Collapse
Affiliation(s)
- Qian Guo
- Laboratory of Drug Delivery, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226019, China
- National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China
| |
Collapse
|
11
|
Lai G, Wu H, Yang K, Hu K, Zhou Y, Chen X, Fu F, Li J, Xie G, Wang HF, Lv Z, Wu X. Progress of nanoparticle drug delivery system for the treatment of glioma. Front Bioeng Biotechnol 2024; 12:1403511. [PMID: 38919382 PMCID: PMC11196769 DOI: 10.3389/fbioe.2024.1403511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/17/2024] [Indexed: 06/27/2024] Open
Abstract
Gliomas are typical malignant brain tumours affecting a wide population worldwide. Operation, as the common treatment for gliomas, is always accompanied by postoperative drug chemotherapy, but cannot cure patients. The main challenges are chemotherapeutic drugs have low blood-brain barrier passage rate and a lot of serious adverse effects, meanwhile, they have difficulty targeting glioma issues. Nowadays, the emergence of nanoparticles (NPs) drug delivery systems (NDDS) has provided a new promising approach for the treatment of gliomas owing to their excellent biodegradability, high stability, good biocompatibility, low toxicity, and minimal adverse effects. Herein, we reviewed the types and delivery mechanisms of NPs currently used in gliomas, including passive and active brain targeting drug delivery. In particular, we primarily focused on various hopeful types of NPs (such as liposome, chitosan, ferritin, graphene oxide, silica nanoparticle, nanogel, neutrophil, and adeno-associated virus), and discussed their advantages, disadvantages, and progress in preclinical trials. Moreover, we outlined the clinical trials of NPs applied in gliomas. According to this review, we provide an outlook of the prospects of NDDS for treating gliomas and summarise some methods that can enhance the targeting specificity and safety of NPs, like surface modification and conjugating ligands and peptides. Although there are still some limitations of these NPs, NDDS will offer the potential for curing glioma patients.
Collapse
Affiliation(s)
- Guogang Lai
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hao Wu
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaixia Yang
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaikai Hu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yan Zhou
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiao Chen
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Fan Fu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiayi Li
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guomin Xie
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hai-Feng Wang
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zhongyue Lv
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiping Wu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
12
|
Huang T, Wen X, Liang Y, Liu X, Zhao J, Long X. Irreversible Electroporation-Induced Inflammation Facilitates Neutrophil-Mediated Drug Delivery to Enhance Pancreatic Cancer Therapy. Mol Pharm 2024; 21:1998-2011. [PMID: 38412284 DOI: 10.1021/acs.molpharmaceut.4c00006] [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: 02/29/2024]
Abstract
Pancreatic cancer is a deadly disease with a five-year overall survival rate of around 11%. Chemotherapy is a cornerstone in the treatment of this malignancy, but the intratumoral delivery of chemotherapy drugs is impaired by the highly fibrotic tumor-associated stroma. Irreversible electroporation (IRE) is an ablative technique for treating locally advanced pancreatic cancer. During a typical IRE procedure, high-intensity electric pulses are released to kill tumor cells through the irreversible disruption of the cytoplasm membranes. IRE also induces rapid tumor infiltration by neutrophils and offers an opportunity for neutrophil-mediated drug delivery. We herein showed that the IRE-induced neutrophil trafficking was facilitated by the upregulation of neutrophil chemotaxis and migration as well as the release of several chemoattractants. Doxorubicin-loaded bovine serum albumin nanoparticles were prepared and loaded into neutrophils at a ratio of 9.9 ± 1.2 to 11.7 ± 2.0 pg of doxorubicin per cell. The resultant formulation (NP@NEs) efficiently accumulated in the IRE-treated KPC-A377 murine pancreatic tumors with an uptake value of 10.7 ± 1.5 (percent of injected dose per gram of tissue, abbreviated as %ID/g) at 48 h after intravenous injection. In both Panc02 and KPC-A377 murine pancreatic tumor models, the combination of IRE + NP@NEs inhibited tumor growth more effectively than either monotherapy. The tumors treated with the combination also exhibited the lowest frequency of Ki67+ proliferating cells and the highest abundance of terminal deoxynucleotidyl transferase dUTP nick end labeling+ (TUNEL+) apoptotic cells among the experiment groups. Minimal treatment-associated toxicity was observed. Our findings suggest that neutrophil-mediated delivery of chemotherapy drugs is a useful tool to enhance the response of pancreatic cancer to IRE.
Collapse
Affiliation(s)
- Teng Huang
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaofei Wen
- Department of Interventional Radiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 36100, China
- Department of Interventional Radiology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Yuxuan Liang
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiao Liu
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jun Zhao
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xin Long
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
13
|
Zhang SS, Li RQ, Chen Z, Wang XY, Dumont AS, Fan X. Immune cells: potential carriers or agents for drug delivery to the central nervous system. Mil Med Res 2024; 11:19. [PMID: 38549161 PMCID: PMC10979586 DOI: 10.1186/s40779-024-00521-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
Drug delivery systems (DDS) have recently emerged as a promising approach for the unique advantages of drug protection and targeted delivery. However, the access of nanoparticles/drugs to the central nervous system (CNS) remains a challenge mainly due to the obstruction from brain barriers. Immune cells infiltrating the CNS in the pathological state have inspired the development of strategies for CNS foundation drug delivery. Herein, we outline the three major brain barriers in the CNS and the mechanisms by which immune cells migrate across the blood-brain barrier. We subsequently review biomimetic strategies utilizing immune cell-based nanoparticles for the delivery of nanoparticles/drugs to the CNS, as well as recent progress in rationally engineering immune cell-based DDS for CNS diseases. Finally, we discuss the challenges and opportunities of immune cell-based DDS in CNS diseases to promote their clinical development.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Ruo-Qi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Zhong Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Xiao-Ying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA.
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
14
|
Tian F, Zhou Y, Ma Z, Tang R, Wang X. Organismal Function Enhancement through Biomaterial Intervention. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:377. [PMID: 38392750 PMCID: PMC10891834 DOI: 10.3390/nano14040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 02/24/2024]
Abstract
Living organisms in nature, such as magnetotactic bacteria and eggs, generate various organic-inorganic hybrid materials, providing unique functionalities. Inspired by such natural hybrid materials, researchers can reasonably integrate biomaterials with living organisms either internally or externally to enhance their inherent capabilities and generate new functionalities. Currently, the approaches to enhancing organismal function through biomaterial intervention have undergone rapid development, progressing from the cellular level to the subcellular or multicellular level. In this review, we will concentrate on three key strategies related to biomaterial-guided bioenhancement, including biointerface engineering, artificial organelles, and 3D multicellular immune niches. For biointerface engineering, excess of amino acid residues on the surfaces of cells or viruses enables the assembly of materials to form versatile artificial shells, facilitating vaccine engineering and biological camouflage. Artificial organelles refer to artificial subcellular reactors made of biomaterials that persist in the cytoplasm, which imparts cells with on-demand regulatory ability. Moreover, macroscale biomaterials with spatiotemporal regulation characters enable the local recruitment and aggregation of cells, denoting multicellular niche to enhance crosstalk between cells and antigens. Collectively, harnessing the programmable chemical and biological attributes of biomaterials for organismal function enhancement shows significant potential in forthcoming biomedical applications.
Collapse
Affiliation(s)
- Fengchao Tian
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Yuemin Zhou
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Zaiqiang Ma
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Ruikang Tang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
| |
Collapse
|
15
|
Sheng S, Jin L, Zhang Y, Sun W, Mei L, Zhu D, Dong X, Lv F. A Twindrive Precise Delivery System of Platelet-Neutrophil Hybrid Membrane Regulates Macrophage Combined with CD47 Blocking for Postoperative Immunotherapy. ACS NANO 2024; 18:4981-4992. [PMID: 38193386 DOI: 10.1021/acsnano.3c10862] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
During wound healing after cancer surgery, platelets, neutrophils, and macrophages accumulate at the wound site and induce important pathophysiological features. Utilizing these pathophysiological features, the development of targeted delivery systems for postoperative tumor immunotherapy is an important strategy. Herein, a twindrive precise delivery system of hybrid membrane combined with CD47 blocking is developed for targeted delivery and targeted regulation to induce postoperative immunotherapy. The precise delivery system consists of IR820-modified platelet-neutrophil hybrid membranes loaded with R848 nanoparticles. Based on the pathological characteristics of platelet aggregation and neutrophil tendency caused by the wound inflammatory microenvironment after tumor surgery, the twindrive delivery system could achieve targeted delivery and targeted regulation of immune drugs to tumor sites. After precise delivery guided by fluorescence imaging, R848 is targeted to reprogram M2 macrophages into M1 macrophages, stimulate dendritic cell maturation as an adjuvant, and then activate T cell immunity. R848 polarization and CD47 blockade together enhanced the phagocytosis function of macrophages, which combined with T cell-mediated cellular immune response to finally effectively inhibit postsurgical tumor recurrence, metastasis, and prolonged survival time. It develops a targeted delivery and regulatory system for cell-specific responses to the pathophysiological features of wound healing for postoperative immunotherapy.
Collapse
Affiliation(s)
- Shupei Sheng
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Limin Jin
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yan Zhang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Weiting Sun
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xia Dong
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| |
Collapse
|
16
|
Li X, Weller S, Clergeaud G, Andresen TL. A versatile method for conjugating lipid nanoparticles on T cells through combination of click chemistry and metabolic glycoengineering. Biotechnol J 2024; 19:e2300339. [PMID: 38178719 DOI: 10.1002/biot.202300339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/06/2023] [Accepted: 12/25/2023] [Indexed: 01/06/2024]
Abstract
Cell-mediated drug delivery by conjugating nanomedicine to the surface of living cells is a promising strategy for enhancing the efficacy of both drug delivery and cell therapy. It exploits the tissue homing properties of the specific cell types to overcome in vivo barriers and forms a drug depot by directly putting the therapeutic payload in target cells. An important concern of developing this system is the method to conjugate nanoparticles on cells. Herein, we developed a bioorthogonal T cell conjugation strategy using SPAAC click chemistry, which allows controllable and highly efficient conjugation without affecting the viability and functions of the cytotoxic T lymphocytes. Azide groups were incorporated on the surface of T cells through metabolic glycoengineering, followed by reacting with dibenzylcyclooctyne (DBCO) modified lipid nanoparticles (LNPs). LNPs can be conjugated to T cells, allowing for the loading of different drug molecules on the cells. The metabolic engineering and click reaction approach provides a simple and versatile strategy to conjugate NPs to living cells and enable the development of sophisticated therapeutic cell products.
Collapse
Affiliation(s)
- Xin Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Sven Weller
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Gael Clergeaud
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
17
|
Ji W, Zhang Y, Deng Y, Li C, Kankala RK, Chen A. Nature-inspired nanocarriers for improving drug therapy of atherosclerosis. Regen Biomater 2023; 10:rbad069. [PMID: 37641591 PMCID: PMC10460486 DOI: 10.1093/rb/rbad069] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/22/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Atherosclerosis (AS) has emerged as one of the prevalent arterial vascular diseases characterized by plaque and inflammation, primarily causing disability and mortality globally. Drug therapy remains the main treatment for AS. However, a series of obstacles hinder effective drug delivery. Nature, from natural micro-/nano-structural biological particles like natural cells and extracellular vesicles to the distinctions between the normal and pathological microenvironment, offers compelling solutions for efficient drug delivery. Nature-inspired nanocarriers of synthetic stimulus-responsive materials and natural components, such as lipids, proteins and membrane structures, have emerged as promising candidates for fulfilling drug delivery needs. These nanocarriers offer several advantages, including prolonged blood circulation, targeted plaque delivery, targeted specific cells delivery and controlled drug release at the action site. In this review, we discuss the nature-inspired nanocarriers which leverage the natural properties of cells or the microenvironment to improve atherosclerotic drug therapy. Finally, we provide an overview of the challenges and opportunities of applying these innovative nature-inspired nanocarriers.
Collapse
Affiliation(s)
- Weihong Ji
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Yuanxing Zhang
- The Institute of Forensic Science, Xiamen Public Security Bureau, Xiamen, Fujian 361104, PR China
| | - Yuanru Deng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Changyong Li
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| |
Collapse
|
18
|
Bader LPE, Klok HA. Chemical Approaches for the Preparation of Bacteria - Nano/Microparticle Hybrid Systems. Macromol Biosci 2023; 23:e2200440. [PMID: 36454518 DOI: 10.1002/mabi.202200440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/24/2022] [Indexed: 12/05/2022]
Abstract
Bacteria represent a class of living cells that are very attractive carriers for the transport and delivery of nano- and microsized particles. The use of cell-based carriers, such as for example bacteria, may allow to precisely direct nano- or microsized cargo to a desired site, which would greatly enhance the selectivity of drug delivery and allow to mitigate side effects. One key step towards the use of such nano-/microparticle - bacteria hybrids is the immobilization of the cargo on the bacterial cell surface. To fabricate bacteria - nano-/microparticle biohybrid microsystems, a wide range of chemical approaches are available that can be used to immobilize the particle payload on the bacterial cell surface. This article presents an overview of the various covalent and noncovalent chemistries that are available for the preparation of bacteria - nano-/microparticle hybrids. For each of the different chemical approaches, an overview will be presented that lists the bacterial strains that have been modified, the type and size of nanoparticles that have been immobilized, as well as the methods that have been used to characterize the nanoparticle-modified bacteria.
Collapse
Affiliation(s)
- Lisa Patricia Elisabeth Bader
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, Lausanne, CH-1015, Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, Lausanne, CH-1015, Switzerland
| |
Collapse
|
19
|
Yue NN, Xu HM, Xu J, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Yao J, Liang YJ, Li DF, Wang LS. Application of Nanoparticles in the Diagnosis of Gastrointestinal Diseases: A Complete Future Perspective. Int J Nanomedicine 2023; 18:4143-4170. [PMID: 37525691 PMCID: PMC10387254 DOI: 10.2147/ijn.s413141] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/02/2023] [Indexed: 08/02/2023] Open
Abstract
The diagnosis of gastrointestinal (GI) diseases currently relies primarily on invasive procedures like digestive endoscopy. However, these procedures can cause discomfort, respiratory issues, and bacterial infections in patients, both during and after the examination. In recent years, nanomedicine has emerged as a promising field, providing significant advancements in diagnostic techniques. Nanoprobes, in particular, offer distinct advantages, such as high specificity and sensitivity in detecting GI diseases. Integration of nanoprobes with advanced imaging techniques, such as nuclear magnetic resonance, optical fluorescence imaging, tomography, and optical correlation tomography, has significantly enhanced the detection capabilities for GI tumors and inflammatory bowel disease (IBD). This synergy enables early diagnosis and precise staging of GI disorders. Among the nanoparticles investigated for clinical applications, superparamagnetic iron oxide, quantum dots, single carbon nanotubes, and nanocages have emerged as extensively studied and utilized agents. This review aimed to provide insights into the potential applications of nanoparticles in modern imaging techniques, with a specific focus on their role in facilitating early and specific diagnosis of a range of GI disorders, including IBD and colorectal cancer (CRC). Additionally, we discussed the challenges associated with the implementation of nanotechnology-based GI diagnostics and explored future prospects for translation in this promising field.
Collapse
Affiliation(s)
- Ning-ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
20
|
Liao X, Gong G, Dai M, Xiang Z, Pan J, He X, Shang J, Blocki AM, Zhao Z, Shields CW, Guo J. Systemic Tumor Suppression via Macrophage-Driven Automated Homing of Metal-Phenolic-Gated Nanosponges for Metastatic Melanoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207488. [PMID: 37072673 PMCID: PMC10288275 DOI: 10.1002/advs.202207488] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/07/2023] [Indexed: 05/03/2023]
Abstract
Cell-based therapies comprising the administration of living cells to patients for direct therapeutic activities have experienced remarkable success in the clinic, of which macrophages hold great potential for targeted drug delivery due to their inherent chemotactic mobility and homing ability to tumors with high efficiency. However, such targeted delivery of drugs through cellular systems remains a significant challenge due to the complexity of balancing high drug-loading with high accumulations in solid tumors. Herein, a tumor-targeting cellular drug delivery system (MAGN) by surface engineering of tumor-homing macrophages (Mφs) with biologically responsive nanosponges is reported. The pores of the nanosponges are blocked with iron-tannic acid complexes that serve as gatekeepers by holding encapsulated drugs until reaching the acidic tumor microenvironment. Molecular dynamics simulations and interfacial force studies are performed to provide mechanistic insights into the "ON-OFF" gating effect of the polyphenol-based supramolecular gatekeepers on the nanosponge channels. The cellular chemotaxis of the Mφ carriers enabled efficient tumor-targeted delivery of drugs and systemic suppression of tumor burden and lung metastases in vivo. The findings suggest that the MAGN platform offers a versatile strategy to efficiently load therapeutic drugs to treat advanced metastatic cancers with a high loading capacity of various therapeutic drugs.
Collapse
Affiliation(s)
- Xue Liao
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Guidong Gong
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
| | - Mengyuan Dai
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Zhenyu Xiang
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Jiezhou Pan
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Xianglian He
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Jiaojiao Shang
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
| | - Anna Maria Blocki
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Illinois at ChicagoChicagoIL60612USA
| | - C. Wyatt Shields
- Department of Chemical and Biological EngineeringUniversity of ColoradoBoulderCO80303USA
| | - Junling Guo
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
- Bioproducts InstituteDepartment of Chemical and Biological EngineeringUniversity of British ColumbiaVancouverBCV6T 1Z4Canada
- State Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengduSichuan610065China
| |
Collapse
|
21
|
Wang T, Yin Q, Huang HY, Wang Z, Song H, Luo X. Probiotic Escherichia coli Nissle 1917 propelled micro-robot with pH sensitivity for hypoxia targeted intestinal tumor therapy. Colloids Surf B Biointerfaces 2023; 225:113277. [PMID: 36996630 DOI: 10.1016/j.colsurfb.2023.113277] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Poor drug penetration in hypoxia area of solid tumor is a big challenge for intestinal tumor therapy and thus it is crucial to develop an effective strategy to overcome this challenge. Compared with other bacteria used for construction of hypoxia targeted bacteria micro-robot, the Escherichia coli Nissle 1917 (EcN) bacteria are nonpathogenic Gram-negative probiotic and can especially target and identify the signal molecules in the hypoxic region of tumor, and thus, in this study, we choose EcN to construct a bacteria propelled micro-robot for targeting intestinal tumor therapy. Firstly, the MSNs@DOX with average diameter of 200 nm were synthesized and conjugated with EcN bacteria using EDC/NHS chemical crosslinking method to construct a EcN propelled micro-robot. The motility of micro-robot was then evaluated and the motion velocity of EcN-pMSNs@DOX was 3.78 µm/s. Compared with pMSNs@DOX without EcN driven, EcN bacteria propelled micro-robot transported much more pMSNs@DOX into the inner of HCT-116 3D multicellular tumor spheroids. However, the EcN bacteria are non-intracelluar bacteria which lead to the micro-robot can not directly enter into tumor cells. Therefore, we utilized acid-labile linkers of cis-aconitic amido bone to link EcN with MSNs@DOX nanoparticles to achieve the pH sensitive separation of EcN with MSNs@DOX from the micro-robot. At 4 h of incubation, the isolated MSNs@DOX began to enter into the tumor cells through CLSM observation. In vitro live/dead staining results show that EcN-pMSNs@DOX induced much more cell death than pMSNs@DOX at 24 and 48 h of incubation with HCT-116 tumor cells in acid culture media (pH 5.3). For the validation of the therapeutic efficacy of the micro-robot for intestinal tumor, we established the HCT-116 subcutaneous transplantation tumor model. After 28 days of treatment, EcN-pMSNs@DOX dramatically inhibit tumor growth with tumor volume was around 689 mm3, induce much more tumor tissues necrosis and apoptosis. Finally, the toxicity of this micro-robot was investigated by pathological analysis the liver and heart tissues. We expect that the pH sensitive EcN propelled micro-robot here we constructed may be a safe and feasible strategy for intestinal tumor therapy.
Collapse
|
22
|
Yu X, Xing G, Sheng S, Jin L, Zhang Y, Zhu D, Mei L, Dong X, Lv F. Neutrophil Camouflaged Stealth Nanovehicle for Photothermal-Induced Tumor Immunotherapy by Triggering Pyroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207456. [PMID: 36967574 DOI: 10.1002/advs.202207456] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/03/2023] [Indexed: 05/27/2023]
Abstract
The regulation of tumor immunosuppressive microenvironments via precise drug delivery is a promising strategy for preventing tumor recurrence and metastasis. Inspired by the stealth strategy, a stealthy nanovehicle based on neutrophil camouflage is developed to achieve precise delivery and tumor immunotherapy by triggering pyroptosis. The nanovehicle comprises anti-CD11b- and IR820-conjugated bovine serum albumin nanoparticles loaded with decitabine. Camouflaged by neutrophils, the nanovehicles achieve efficient tumor delivery by neutrophil hitchhiking owing to the biotropism of neutrophils for tumors. The fluorescent signal molecule, IR820, on the nanovehicle acts as a navigation monitor to track the precise delivery of the nanovehicle. The released decitabine upregulates gasdermin E, and laser irradiation activates caspase-3, thereby resulting in pyroptosis, which improves the system's adaptive immune response. In a triple-negative breast cancer animal model, it regulates the immunosuppressive microenvironment for effective tumor immunotherapy and induces a long-lasting and strong immune memory to prevent lung metastasis.
Collapse
Affiliation(s)
- Xuya Yu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Guozheng Xing
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Shupei Sheng
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Limin Jin
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Yan Zhang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Xia Dong
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P.R. China
| |
Collapse
|
23
|
Zhu K, Xu Y, Zhong R, Li W, Wang H, Wong YS, Venkatraman S, Liu J, Cao Y. Hybrid liposome-erythrocyte drug delivery system for tumor therapy with enhanced targeting and blood circulation. Regen Biomater 2023; 10:rbad045. [PMID: 37250975 PMCID: PMC10224802 DOI: 10.1093/rb/rbad045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/26/2023] [Accepted: 04/11/2023] [Indexed: 05/31/2023] Open
Abstract
Liposome, a widely used drug delivery system (DDS), still shows several disadvantages such as dominant clearance by liver and poor target organ deposition. To overcome the drawbacks of liposomes, we developed a novel red blood cell (RBC)-liposome combined DDS to modulate the tumor accumulation and extend the blood circulation life of the existing liposomal DDS. Here, RBCs, an ideal natural carrier DDS, were utilized to carry liposomes and avoid them undergo the fast clearance in the blood. In this study, liposomes could either absorbed onto RBCs' surface or fuse with RBCs' membrane by merely altering the interaction time at 37°C, while the interaction between liposome and RBCs would not affect RBCs' characteristics. In the in vivo antitumor therapeutic efficacy study, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) liposomes attached onto RBCs' surfaces exhibited lung targeting effect (via RBC-hitchhiking approach) and reduced clearance in the liver, while DPPC liposomes fused with RBCs had prolong blood circulation up to 48 h and no enrichment in any organ. Furthermore, 20 mol% of DPPC liposomes were replaced with pH-sensitive phospholipid 1,2-dioleoyl-Sn-glycero-3-phosphoethanolamine (DOPE) as it could respond to the low pH tumor microenvironment and then accumulate in the tumor. The DOPE attached/fusion RBCs showed partial enrichment in lung and about 5-8% tumor accumulation, which were significantly higher than (about 0.7%) the conventional liposomal DDS. Thus, RBC-liposome composite DDS is able to improve the liposomal tumor accumulation and blood circulation and shows the clinical application promises of using autologous RBCs for antitumor therapy.
Collapse
Affiliation(s)
- Kehui Zhu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Yingcan Xu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Rui Zhong
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Wanjing Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Hong Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, China
| | - Yee Shan Wong
- Biomedical Engineering, School of Engineering, Temasek Polytechnic, Singapore, Singapore
| | - Subramanian Venkatraman
- School of Materials Science and Engineering, National University of Singapore, Singapore, Singapore
| | - Jiaxin Liu
- Correspondence address. E-mail: (J.L.); , (Y.C.)
| | - Ye Cao
- Correspondence address. E-mail: (J.L.); , (Y.C.)
| |
Collapse
|
24
|
Chen Y, Qin D, Zou J, Li X, Guo XD, Tang Y, Liu C, Chen W, Kong N, Zhang CY, Tao W. Living Leukocyte-Based Drug Delivery Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207787. [PMID: 36317596 DOI: 10.1002/adma.202207787] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/10/2022] [Indexed: 05/17/2023]
Abstract
Leukocytes play a vital role in immune responses, including defending against invasive pathogens, reconstructing impaired tissue, and maintaining immune homeostasis. When the immune system is activated in vivo, leukocytes accomplish a series of orderly and complex regulatory processes. While cancer and inflammation-related diseases like sepsis are critical medical difficulties plaguing humankind around the world, leukocytes have been shown to largely gather at the focal site, and significantly contribute to inflammation and cancer progression. Therefore, the living leukocyte-based drug delivery systems have attracted considerable attention in recent years due to the innate and specific targeting effect, low immunogenicity, improved therapeutic efficacy, and low reverse effect. In this review, the recent advances in the development of living leukocyte-based drug delivery systems including macrophages, neutrophils, and lymphocytes as promising treatment strategies for cancer and inflammation-related diseases are introduced. The advantages, current challenges, and limitations of these delivery systems are also discussed, as well as perspectives on the future development of precision and targeted therapy in the clinics are provided. Collectively, it is expected that such kind of living cell-based drug delivery system is promising to improve or even revolutionize the treatments of cancers and inflammation-related diseases in the clinics.
Collapse
Affiliation(s)
- Yaxin Chen
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Duotian Qin
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jianhua Zou
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR), 519020, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xiaobin Li
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Xin Dong Guo
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yi Tang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Chuang Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Na Kong
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, 311121, China
| | - Can Yang Zhang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 440300, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
25
|
Pawar B, Vasdev N, Gupta T, Mhatre M, More A, Anup N, Tekade RK. Current Update on Transcellular Brain Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14122719. [PMID: 36559214 PMCID: PMC9786068 DOI: 10.3390/pharmaceutics14122719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
It is well known that the presence of a blood-brain barrier (BBB) makes drug delivery to the brain more challenging. There are various mechanistic routes through which therapeutic molecules travel and deliver the drug across the BBB. Among all the routes, the transcellular route is widely explored to deliver therapeutics. Advances in nanotechnology have encouraged scientists to develop novel formulations for brain drug delivery. In this article, we have broadly discussed the BBB as a limitation for brain drug delivery and ways to solve it using novel techniques such as nanomedicine, nose-to-brain drug delivery, and peptide as a drug delivery carrier. In addition, the article will help to understand the different factors governing the permeability of the BBB, as well as various formulation-related factors and the body clearance of the drug delivered into the brain.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rakesh Kumar Tekade
- Correspondence: ; Tel.: +91-796674550 or +91-7966745555; Fax: +91-7966745560
| |
Collapse
|
26
|
Shen N, Qi X, Bagrov DV, Krechetov SP, Sharapov MG, Durymanov MO. Surface modification of fibroblasts with peroxiredoxin-1-loaded polymeric microparticles increases cell mobility, resistance to oxidative stress and collagen I production. Colloids Surf B Biointerfaces 2022; 219:112834. [PMID: 36152599 DOI: 10.1016/j.colsurfb.2022.112834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/20/2022]
Abstract
Modification of the cell surface with artificial nano- and microparticles (also termed "cellular backpacks") containing biologically active payloads usually enables drug targeting via harnessing intrinsic cell tropism to the sites of injury. In some cases, using cells as delivery vehicles leads to improved pharmacokinetics due to extended circulation time of cell-immobilized formulations. Another rationale for particle attachment to cells is augmentation of desirable cellular functions and cell proliferation in response to release of the particle contents. In this study, we conjugated poly(lactic-co-glycolic acid) (PLGA) microparticles loaded with multifunctional antioxidant enzyme peroxiredoxin-1 (Prx1) to the surface of fibroblasts. The obtained microparticles were uniform in size and demonstrated sustained protein release. We found that the released Prx1 maintains its signaling activity resulting in macrophage activation, as indicated by TNFα upregulation and increase in ROS generation. Functionalization of fibroblasts with PLGA/Prx1 microparticles via EDC/sulfo-NHS coupling reaction did not affect cell viability but increased cell migratory properties and collagen I production. Moreover, PLGA/Prx1 backpacks increased resistance of fibroblasts to oxidative stress and attenuated cell senescence. In summary, we have developed a novel approach of fibroblast modification to augment their biological properties, which can be desirable for wound repair, cosmetic dermatology, and tissue engineering.
Collapse
Affiliation(s)
- Ningfei Shen
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
| | - Xiaoli Qi
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
| | - Dmitry V Bagrov
- Faculty of Biology, Moscow State University, Moscow, Russia; Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Sergey P Krechetov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov, Moscow, Russia
| | - Mars G Sharapov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino, Russia
| | - Mikhail O Durymanov
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia.
| |
Collapse
|
27
|
Cai J, Peng J, Zang X, Feng J, Li R, Ren P, Zheng B, Wang J, Wang J, Yan M, Liu J, Deng R, Wang D. Mammary Leukocyte-Assisted Nanoparticle Transport Enhances Targeted Milk Trace Mineral Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200841. [PMID: 35773238 PMCID: PMC9475556 DOI: 10.1002/advs.202200841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/18/2022] [Indexed: 06/15/2023]
Abstract
Nanoparticles are applied as versatile platforms for drug/gene delivery in many applications owing to their long-retention and specific targeting properties in living bodies. However, the delivery mechanism and the beneficial effect of nanoparticle-retention in many organisms remain largely uncertain. Here, the transport and metabolism of mineral nanoparticles in mammary gland during lactation are explored. It is shown that maternal intravenous administration of iron oxide nanoparticles (IONPs; diameter: ≈11.0 nm, surface charge: -29.1 mV, surface area: 1.05 m2 g-1 ) provides elevated iron delivery to mammary gland and increased iron secretion into breast milk, which is inaccessible by classical iron-ion transport approaches such as the transferrin receptor-mediated endocytic pathway. Mammary macrophages and neutrophils are found to play dominant roles in uptake and delivery of IONPs through an unconventional leukocyte-assisted iron secretion pathway. This pathway bypasses the tight iron concentration regulation of liver hepcidin-ferroportin axis and mammary epithelial cells to increase milk iron-ion content derived from IONPs. This work provides keen insight into the metabolic pathway of nanoparticles in mammary gland while offering a new scheme of nutrient delivery for neonate metabolism regulation by using nanosized nutrients.
Collapse
Affiliation(s)
- Jie Cai
- Institute of Dairy ScienceCollege of Animal SciencesMOE Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhou310029P. R. China
| | - Jie Peng
- School of Materials Science and EngineeringState Key Laboratory of Silicon MaterialsInstitute for Composites Science InnovationZhejiang UniversityHangzhou310027P. R. China
| | - Xinwei Zang
- Institute of Dairy ScienceCollege of Animal SciencesMOE Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhou310029P. R. China
| | - Juan Feng
- Institute of Dairy ScienceCollege of Animal SciencesMOE Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhou310029P. R. China
| | - Ruocheng Li
- Institute of Dairy ScienceCollege of Animal SciencesMOE Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhou310029P. R. China
| | - Peng Ren
- Institute of Dairy ScienceCollege of Animal SciencesMOE Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhou310029P. R. China
| | - Bingzhu Zheng
- School of Materials Science and EngineeringState Key Laboratory of Silicon MaterialsInstitute for Composites Science InnovationZhejiang UniversityHangzhou310027P. R. China
| | - Jiaying Wang
- Institute of Environmental HealthMOE Key Laboratory of Environmental Remediation and Ecosystem HealthCollege of Environmental & Resource SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Juan Wang
- Institute of Environmental HealthMOE Key Laboratory of Environmental Remediation and Ecosystem HealthCollege of Environmental & Resource SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Mi Yan
- School of Materials Science and EngineeringState Key Laboratory of Silicon MaterialsInstitute for Composites Science InnovationZhejiang UniversityHangzhou310027P. R. China
| | - Jianxin Liu
- Institute of Dairy ScienceCollege of Animal SciencesMOE Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhou310029P. R. China
| | - Renren Deng
- School of Materials Science and EngineeringState Key Laboratory of Silicon MaterialsInstitute for Composites Science InnovationZhejiang UniversityHangzhou310027P. R. China
- Department of Medical OncologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310003P. R. China
| | - Diming Wang
- Institute of Dairy ScienceCollege of Animal SciencesMOE Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhou310029P. R. China
| |
Collapse
|
28
|
Li X, Halldórsdóttir HR, Weller S, Colliander A, Bak M, Kempen P, Clergeaud G, Andresen TL. Enhancing Adoptive Cell Therapy by T Cell Loading of SHP2 Inhibitor Nanocrystals before Infusion. ACS NANO 2022; 16:10918-10930. [PMID: 35838499 DOI: 10.1021/acsnano.2c03311] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Whereas adoptive T cell therapy has been extensively studied for cancer treatment, the response is still limited primarily due to immune dysfunction related to poor cell engraftment, tumor infiltration and engagement, and lack of a target. In addition, the modification of therapeutic T cells often suffers from being complex and expensive. Here, we present a strategy to load T cells with SHP099, an allosteric SHP2 inhibitor, to enhance the therapeutic efficacy of the T cells. Remote-loading of SHP099 into lipid nanoparticles decorated with triarginine motifs resulted in nanocrystal formation of SHP099 inside the lipid vesicles and allowed high loading efficiency and prolonged retention of SHP099 nanocrystals within T cells. Cell-loaded SHP099 enabled sustained inhibition of the PD-1/PD-L1 signaling and increased cytolytic activity of the T cells. We show in a mouse model that tumor-homing T cells can circulate with the cargos, improving their tumor accumulation compared to systemically administered lipid nanoparticles. On an established solid tumor model, adoptively transferred SHP099 loaded T cells induced complete tumor eradication and durable immune memory against tumor rechallenging on all treated mice by effectively inhibiting the PD-1/PD-L1 checkpoint signal. We demonstrate that the combination of T cell therapy with SHP2 inhibition is a promising therapeutic strategy, and the lipid nanocrystal platform could be generalized as a promising approach for T cell loading of immunomodulatory drugs.
Collapse
Affiliation(s)
- Xin Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | | | - Sven Weller
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Anna Colliander
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Martin Bak
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Paul Kempen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- National Centre for Nano Fabrication and Characterization, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Gael Clergeaud
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
29
|
Su N, Villicana C, Yang F. Immunomodulatory strategies for bone regeneration: A review from the perspective of disease types. Biomaterials 2022; 286:121604. [PMID: 35667249 PMCID: PMC9881498 DOI: 10.1016/j.biomaterials.2022.121604] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 02/08/2023]
Abstract
Tissue engineering strategies for treating bone loss to date have largely focused on targeting stem cells or vascularization. Immune cells, including macrophages and T cells, can also indirectly enhance bone healing via cytokine secretion to interact with other bone niche cells. Bone niche cues and local immune environment vary depending on anatomical location, size of defects and disease types. As such, it is critical to evaluate the role of the immune system in the context of specific bone niche and different disease types. This review focuses on immunomodulation research for bone applications using biomaterials and cell-based strategies, with a unique perspective from different disease types. We first reviewed applications for prolonging orthopaedic implant lifetime and enhancing fracture healing, two clinical challenges where immunomodulatory strategies were initially developed for orthopedic applications. We then reviewed recent research progress in harnessing immunomodulatory strategies for regenerating critical-sized, long bone or cranial bone defects, and treating osteolytic bone diseases. Remaining gaps in knowledge, future directions and opportunities were also discussed.
Collapse
Affiliation(s)
- Ni Su
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Cassandra Villicana
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA.,: Corresponding Author Fan Yang, Ph D, Department of Orthopaedic Surgery and Bioengineering, Stanford University School of Medicine, 240 Pasteur Dr, Palo Alto, CA 94304, Biomedical Innovation Building, 1st floor, Room 1200, , Phone: (650) 646-8558
| |
Collapse
|
30
|
Wang T, Qin J, Cheng J, Li C, Du J. Intelligent design of polymersomes for antibacterial and anticancer applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1822. [PMID: 35673991 DOI: 10.1002/wnan.1822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 01/25/2023]
Abstract
Polymersomes (or polymer vesicles) have attracted much attention for biomedical applications in recent years because their lumen can be used for drug delivery and their coronas and membrane can be modified with a variety of functional groups. Thus, polymersomes are very suitable for improved antibacterial and anticancer therapy. This review mainly highlighted recent advances in the synthetic protocols and design principles of intelligent antibacterial and anticancer polymersomes. Antibacterial polymersomes are divided into three categories: polymersomes as antibiotic nanocarriers, intrinsically antibacterial polymersomes, and antibacterial polymersomes with supplementary means including photothermal and photodynamic therapy. Similarly, the anticancer polymersomes are divided into two categories: polymersomes-based delivery systems and anticancer polymersomes with supplementary means. In addition, the bilateral relationship between bacteria and cancer is addressed, since more and more evidences show that bacteria may cause cancer or promote cancer progression. Finally, prospective on next-generation antibacterial and anticancer polymersomes are discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Tao Wang
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jinlong Qin
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiajing Cheng
- Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
31
|
Grimaldi M, Santoro A, Buonocore M, Crivaro C, Funicello N, Sublimi Saponetti M, Ripoli C, Rodriquez M, De Pasquale S, Bobba F, Ferrazzano L, Cabri W, D’Ursi AM, Ricci A. A New Approach to Supramolecular Structure Determination in Pharmaceutical Preparation of Self-Assembling Peptides: A Case Study of Lanreotide Autogel. Pharmaceutics 2022; 14:pharmaceutics14030681. [PMID: 35336055 PMCID: PMC8954372 DOI: 10.3390/pharmaceutics14030681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 12/13/2022] Open
Abstract
The supramolecular structure in peptides’ prolonged-released gel formulations is the most critical parameter for the determination of the pharmaceutical profile of the drug. Here, we report our investigation on lanreotide Autogel as a case study. For the first time, we describe the use of the pulsed field gradient (PFG) diffusion-ordered spectroscopy (DOSY) magic-angle spinning NMR to characterize the supramolecular self-assembly and molecular mobility of different samples of lanreotide Autogel formulations prepared according to different formulation protocols. The diffusion coefficient was used to calculate the hydrodynamic radii of supramolecular assemblies and build relative molecular models. DOSY data were integrated with NMR imaging (MRI) measurements and atomic force microscopy (AFM) imaging.
Collapse
Affiliation(s)
- Manuela Grimaldi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Salerno, Italy; (M.G.); (A.S.); (M.B.); (M.R.)
| | - Angelo Santoro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Salerno, Italy; (M.G.); (A.S.); (M.B.); (M.R.)
| | - Michela Buonocore
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Salerno, Italy; (M.G.); (A.S.); (M.B.); (M.R.)
| | - Claudio Crivaro
- Fresenius Kabi iPSUM, Via San Leonardo 23, 45010 Villadose, Italy; (C.C.); (A.R.)
| | - Nicola Funicello
- Department of Physics ‘E.R. Caianiello’ of University and Gruppo Collegato INFN, 84084 Salerno, Italy; (N.F.); (C.R.); (S.D.P.)
| | - Matilde Sublimi Saponetti
- Physics Department and Research Centre for Nanomaterials and Nanotechnology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy; (M.S.S.); (F.B.)
| | - Cristina Ripoli
- Department of Physics ‘E.R. Caianiello’ of University and Gruppo Collegato INFN, 84084 Salerno, Italy; (N.F.); (C.R.); (S.D.P.)
| | - Manuela Rodriquez
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Salerno, Italy; (M.G.); (A.S.); (M.B.); (M.R.)
| | - Salvatore De Pasquale
- Department of Physics ‘E.R. Caianiello’ of University and Gruppo Collegato INFN, 84084 Salerno, Italy; (N.F.); (C.R.); (S.D.P.)
| | - Fabrizio Bobba
- Physics Department and Research Centre for Nanomaterials and Nanotechnology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy; (M.S.S.); (F.B.)
| | - Lucia Ferrazzano
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum, University of Bologna, Via Selmi 2, 40126 Bologna, Italy;
| | - Walter Cabri
- Fresenius Kabi iPSUM, Via San Leonardo 23, 45010 Villadose, Italy; (C.C.); (A.R.)
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum, University of Bologna, Via Selmi 2, 40126 Bologna, Italy;
- Correspondence: (W.C.); (A.M.D.); Tel.: +39-08996-9748 (A.M.D.)
| | - Anna Maria D’Ursi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Salerno, Italy; (M.G.); (A.S.); (M.B.); (M.R.)
- Correspondence: (W.C.); (A.M.D.); Tel.: +39-08996-9748 (A.M.D.)
| | - Antonio Ricci
- Fresenius Kabi iPSUM, Via San Leonardo 23, 45010 Villadose, Italy; (C.C.); (A.R.)
| |
Collapse
|
32
|
Cui J, Xu Y, Tu H, Zhao H, Wang H, Di L, Wang R. Gather wisdom to overcome barriers: Well-designed nano-drug delivery systems for treating gliomas. Acta Pharm Sin B 2022; 12:1100-1125. [PMID: 35530155 PMCID: PMC9069319 DOI: 10.1016/j.apsb.2021.08.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/07/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Due to the special physiological and pathological characteristics of gliomas, most therapeutic drugs are prevented from entering the brain. To improve the poor prognosis of existing therapies, researchers have been continuously developing non-invasive methods to overcome barriers to gliomas therapy. Although these strategies can be used clinically to overcome the blood‒brain barrier (BBB), the accurate delivery of drugs to the glioma lesions cannot be ensured. Nano-drug delivery systems (NDDS) have been widely used for precise drug delivery. In recent years, researchers have gathered their wisdom to overcome barriers, so many well-designed NDDS have performed prominently in preclinical studies. These meticulous designs mainly include cascade passing through BBB and targeting to glioma lesions, drug release in response to the glioma microenvironment, biomimetic delivery systems based on endogenous cells/extracellular vesicles/protein, and carriers created according to the active ingredients of traditional Chinese medicines. We reviewed these well-designed NDDS in detail. Furthermore, we discussed the current ongoing and completed clinical trials of NDDS for gliomas therapy, and analyzed the challenges and trends faced by clinical translation of these well-designed NDDS.
Collapse
Affiliation(s)
- Jiwei Cui
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology, Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yuanxin Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology, Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Haiyan Tu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology, Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Huacong Zhao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology, Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Honglan Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology, Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology, Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Ruoning Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology, Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
- Corresponding author. Tel./fax: +86 15852937869.
| |
Collapse
|
33
|
Ding X, Sun X, Cai H, Wu L, Liu Y, Zhao Y, Zhou D, Yu G, Zhou X. Engineering Macrophages via Nanotechnology and Genetic Manipulation for Cancer Therapy. Front Oncol 2022; 11:786913. [PMID: 35070992 PMCID: PMC8770285 DOI: 10.3389/fonc.2021.786913] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages play critical roles in tumor progression. In the tumor microenvironment, macrophages display highly diverse phenotypes and may perform antitumorigenic or protumorigenic functions in a context-dependent manner. Recent studies have shown that macrophages can be engineered to transport drug nanoparticles (NPs) to tumor sites in a targeted manner, thereby exerting significant anticancer effects. In addition, macrophages engineered to express chimeric antigen receptors (CARs) were shown to actively migrate to tumor sites and eliminate tumor cells through phagocytosis. Importantly, after reaching tumor sites, these engineered macrophages can significantly change the otherwise immune-suppressive tumor microenvironment and thereby enhance T cell-mediated anticancer immune responses. In this review, we first introduce the multifaceted activities of macrophages and the principles of nanotechnology in cancer therapy and then elaborate on macrophage engineering via nanotechnology or genetic approaches and discuss the effects, mechanisms, and limitations of such engineered macrophages, with a focus on using live macrophages as carriers to actively deliver NP drugs to tumor sites. Several new directions in macrophage engineering are reviewed, such as transporting NP drugs through macrophage cell membranes or extracellular vesicles, reprogramming tumor-associated macrophages (TAMs) by nanotechnology, and engineering macrophages with CARs. Finally, we discuss the possibility of combining engineered macrophages and other treatments to improve outcomes in cancer therapy.
Collapse
Affiliation(s)
- Xiaoling Ding
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xinchen Sun
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, Taizhou Peoples' Hospital, Taizhou, China
| | - Huihui Cai
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, The Sixth Nantong People's Hospital, Nantong, China
| | - Lei Wu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Ying Liu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yu Zhao
- Department of Immunology, Southeast University, School of Medicine, Nanjing, China
| | - Dingjingyu Zhou
- Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China
| | - Xiaorong Zhou
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| |
Collapse
|
34
|
Ding L, Zhang P, Huang X, Yang K, Liu X, Yu Z. Intracellular Reduction-Responsive Molecular Targeted Nanomedicine for Hepatocellular Carcinoma Therapy. Front Pharmacol 2022; 12:809125. [PMID: 35082681 PMCID: PMC8784786 DOI: 10.3389/fphar.2021.809125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/30/2021] [Indexed: 01/20/2023] Open
Abstract
The stimuli-responsive polymer-based platform for controlled drug delivery has gained increasing attention in treating hepatocellular carcinoma (HCC) owing to the fascinating biocompatibility and biodegradability, improved antitumor efficacy, and negligible side effects recently. Herein, a disulfide bond-contained polypeptide nanogel, methoxy poly(ethylene glycol)-poly(l-phenylalanine-co-l-cystine) [mPEG-P(LP-co-LC)] nanogel, which could be responsive to the intracellular reduction microenvironments, was developed to deliver lenvatinib (LEN), an inhibitor of multiple receptor tyrosine kinases, for HCC therapy. The lenvatinib-loaded nanogel (NG/LEN) displayed concise drug delivery under the stimulus of glutathione in the cancer cells. Furthermore, the intracellular reduction-responsive nanomedicine NG/LEN showed excellent antitumor effect and almost no side effects toward both subcutaneous and orthotopic HCC tumor-allografted mice in comparison to free drug. The excellent tumor-inhibition efficacy with negligible side effects demonstrated the potential of NG/LEN for clinical molecular targeted therapy of gastrointestinal carcinoma in the future.
Collapse
Affiliation(s)
- Lei Ding
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xu Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Kunmeng Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xingkai Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Zhenxiang Yu
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
35
|
Tang L, He S, Yin Y, Liu H, Hu J, Cheng J, Wang W. Combination of Nanomaterials in Cell-Based Drug Delivery Systems for Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13111888. [PMID: 34834304 PMCID: PMC8621332 DOI: 10.3390/pharmaceutics13111888] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cell-based drug delivery systems have shown tremendous advantages in cancer treatment due to their distinctive properties. For instance, delivery of therapeutics using tumor-tropic cells like neutrophils, lymphocytes and mesenchymal stem cells can achieve specific tumor targeting due to the "Trojan Horse" effect. Other circulatory cells like erythrocytes and platelets can greatly improve the circulation time of nanoparticles due to their innate long circulation property. Adipocytes, especially cancer-associated adipocytes, play key roles in tumor development and metabolism, therefore, adipocytes are regarded as promising bio-derived nanoplatforms for anticancer targeted drug delivery. Nanomaterials are important participants in cell-based drug delivery because of their unique physicochemical characteristics. Therefore, the integration of various nanomaterials with different cell types will endow the constructed delivery systems with many attractive properties due to the merits of both. In this review, a number of strategies based on nanomaterial-involved cell-mediated drug delivery systems for cancer treatment will be summarized. This review discusses how nanomaterials can be a benefit to cell-based therapies and how cell-derived carriers overcome the limitations of nanomaterials, which highlights recent advancements and specific biomedical applications based on nanomaterial-mediated, cell-based drug delivery systems.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Jingyi Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 211166, China
- Correspondence: (J.C.); (W.W.)
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (L.T.); (S.H.); (Y.Y.); (H.L.); (J.H.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 211198, China
- Correspondence: (J.C.); (W.W.)
| |
Collapse
|
36
|
Battistella C, Liang Y, Gianneschi NC. Innovations in Disease State Responsive Soft Materials for Targeting Extracellular Stimuli Associated with Cancer, Cardiovascular Disease, Diabetes, and Beyond. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007504. [PMID: 34145625 PMCID: PMC9836048 DOI: 10.1002/adma.202007504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/20/2021] [Indexed: 05/10/2023]
Abstract
Recent advances in polymer chemistry, materials sciences, and biotechnology have allowed the preclinical development of sophisticated programmable nanomedicines and materials that are able to precisely respond to specific disease-associated triggers and microenvironments. These stimuli, endogenous to the targeted diseases, include pH, redox-state, small molecules, and protein upregulation. Herein, recent advances and innovative approaches in programmable soft materials capable of sensing the aforementioned disease-associated stimuli and responding via a range of dynamic processes including morphological and size transitions, changes in mobility and retention, as well as disassembly are described. In this field generally, the majority of ongoing and past research effort has focused on oncology. Given this interest, examples of the latest innovative approaches to chemo- and immunotherapy treatment strategies for cancer are presented. Moreover, as the field broadens its attention, applications of programmable materials in other diseases are highlighted, with a special focus on cardiovascular disease and diabetes mellitus, where limited attention is paid by the field, but where many promising avenues exist with high potential impact.
Collapse
Affiliation(s)
- Claudia Battistella
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Department of Pharmacology, Northwestern University, Evanston, IL, 60208, USA
| | - Yifei Liang
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Nathan C Gianneschi
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Department of Pharmacology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
37
|
Wu Y, Han X, Zheng R, Cheng H, Yan J, Wu X, Hu Y, Li B, Wang Z, Li X, Zhang H. Neutrophil mediated postoperative photoimmunotherapy against melanoma skin cancer. NANOSCALE 2021; 13:14825-14836. [PMID: 34533171 DOI: 10.1039/d1nr04002b] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Surgery is the primary treatment option for most melanoma; however, high tumor recurrence rate after surgical resection becomes the main cause of death in cancer patients. The development of efficient drug delivery nanosystems to inhibit postoperative tumor recurrence becomes very necessary. In the present study, IR780 molecules and TRP-2 peptide were encapsulated in the hydrophobic shell and hydrophilic interior of TAT peptide functionalized liposomes to form TLipIT NPs, which were further internalized into neutrophils (NEs) to achieve TLipIT/NEs. After being intravenously injected into postoperative B16F10-bearing mice, TLipIT/NEs could actively migrate toward the inflamed residual tumor and release TLipIT through neutrophil extracellular traps (NETs). Under NIR laser irradiation, the TLipIT exhibited both photothermal and photodynamic effects to induce immunogenic cell death for maturation of DCs, and simultaneously, to release TRP-2 peptide as a melanoma associated antigen to further strengthen the maturation of DCs, both of which prompts the activation of T cells and induces potent immune responses. TLipIT/NEs hold great potential for the inhibition of postoperative tumor recurrence.
Collapse
Affiliation(s)
- Yunyun Wu
- School of Chemistry and Life Science, Changchun University of Technology, Changchun 130012, P.R. China
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China.
| | - Xiaoqing Han
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China.
| | - Runxiao Zheng
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China.
| | - Hongda Cheng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China
| | - Jiao Yan
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China.
| | - Xiaqing Wu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China.
| | - Yaqing Hu
- School of Chemistry and Life Science, Changchun University of Technology, Changchun 130012, P.R. China
| | - Bing Li
- School of Chemistry and Life Science, Changchun University of Technology, Changchun 130012, P.R. China
| | - Zhenxin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xi Li
- School of Chemistry and Life Science, Changchun University of Technology, Changchun 130012, P.R. China
| | - Haiyuan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, 130022, China.
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
38
|
Wang F, Hou W, Xiao C, Hao Y, Su N, Deng Y, Wang J, Yu L, Xie JM, Xiong JW, Luo Y. Endothelial cell membrane-based biosurface for targeted delivery to acute injury: analysis of leukocyte-mediated nanoparticle transportation. NANOSCALE 2021; 13:14636-14643. [PMID: 34558568 DOI: 10.1039/d1nr04181a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Mimicking and leveraging biological structures and materials provide important approaches to develop functional vehicles for drug delivery. Taking advantage of the affinity and adhesion between the activated endothelial cells and innate immune cells during inflammatory responses, hybrid polyester nanoparticles coated with endothelial cell membranes (EM-P) containing adhesion molecules were fabricated and their capability as vehicles to travel to the acute injury sites through leukocyte-mediated processes was investigated. The in vivo studies and quantitative analyses performed through the lung-inflammation mouse models demonstrated that the EM-Ps preferentially interacted with the neutrophils and monocytes in the circulation and the cellular membrane-based biosurface improved the nanoparticle transportation to the inflamed lung possibly via the motility of neutrophils. Utilizing the transgenic zebrafish model, the leukocyte-mediated transportation and biodistribution of EM-Ps were further visualized in real time at the whole-organism level. Endothelial membranes provided a new biosurface for developing biomimetic vehicles to allow the immune cell-mediated transportation and may enable advanced systems for active and highly efficient drug delivery.
Collapse
Affiliation(s)
- Fang Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Wenda Hou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Chenglu Xiao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Yaoyao Hao
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Ni Su
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Yu Deng
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jieting Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Luying Yu
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jing-Ming Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America 02155
| |
Collapse
|
39
|
Qi J, Jin F, You Y, Du Y, Liu D, Xu X, Wang J, Zhu L, Chen M, Shu G, Wu L, Ji J, Du Y. Synergistic effect of tumor chemo-immunotherapy induced by leukocyte-hitchhiking thermal-sensitive micelles. Nat Commun 2021; 12:4755. [PMID: 34362890 PMCID: PMC8346467 DOI: 10.1038/s41467-021-24902-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/14/2021] [Indexed: 01/04/2023] Open
Abstract
Some specific chemotherapeutic drugs are able to enhance tumor immunogenicity and facilitate antitumor immunity by inducing immunogenic cell death (ICD). However, tumor immunosuppression induced by the adenosine pathway hampers this effect. In this study, E-selectin-modified thermal-sensitive micelles are designed to co-deliver a chemotherapeutic drug (doxorubicin, DOX) and an A2A adenosine receptor antagonist (SCH 58261), which simultaneously exhibit chemo-immunotherapeutic effects when applied with microwave irradiation. After intravenous injection, the fabricated micelles effectively adhere to the surface of leukocytes in peripheral blood mediated by E-selectin, and thereby hitchhiking with leukocytes to achieve a higher accumulation at the tumor site. Further, local microwave irradiation is applied to induce hyperthermia and accelerates the release rate of drugs from micelles. Rapidly released DOX induces tumor ICD and elicits tumor-specific immunity, while SCH 58261 alleviates immunosuppression caused by the adenosine pathway, further enhancing DOX-induced antitumor immunity. In conclusion, this study presents a strategy to increase the tumor accumulation of drugs by hitchhiking with leukocytes, and the synergistic strategy of chemo-immunotherapy not only effectively arrested primary tumor growth, but also exhibited superior effects in terms of antimetastasis, antirecurrence and antirechallenge. Targeting the adenosinergic pathway represents a therapeutic option to overcome tumor-induced immunosuppression. Here the authors design E-selectin-modified thermal-sensitive micelles loaded with doxorubicin and an adenosine A2 receptor antagonist to enhance chemotherapy-induced anti-tumor immune responses.
Collapse
Affiliation(s)
- Jing Qi
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feiyang Jin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuchan You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yan Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Di Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Jun Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Minjiang Chen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Gaofeng Shu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, Lishui, China
| | - Liming Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, Lishui, China.
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
40
|
Shi Y, Lu A, Wang X, Belhadj Z, Wang J, Zhang Q. A review of existing strategies for designing long-acting parenteral formulations: Focus on underlying mechanisms, and future perspectives. Acta Pharm Sin B 2021; 11:2396-2415. [PMID: 34522592 PMCID: PMC8424287 DOI: 10.1016/j.apsb.2021.05.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/03/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
The need for long-term treatments of chronic diseases has motivated the widespread development of long-acting parenteral formulations (LAPFs) with the aim of improving drug pharmacokinetics and therapeutic efficacy. LAPFs have been proven to extend the half-life of therapeutics, as well as to improve patient adherence; consequently, this enhances the outcome of therapy positively. Over past decades, considerable progress has been made in designing effective LAPFs in both preclinical and clinical settings. Here we review the latest advances of LAPFs in preclinical and clinical stages, focusing on the strategies and underlying mechanisms for achieving long acting. Existing strategies are classified into manipulation of in vivo clearance and manipulation of drug release from delivery systems, respectively. And the current challenges and prospects of each strategy are discussed. In addition, we also briefly discuss the design principles of LAPFs and provide future perspectives of the rational design of more effective LAPFs for their further clinical translation.
Collapse
Key Words
- 2′-F, 2′-fluoro
- 2′-O-MOE, 2′-O-(2-methoxyethyl)
- 2′-OMe, 2′-O-methyl
- 3D, three-dimensional
- ART, antiretroviral therapy
- ASO, antisense oligonucleotide
- Biomimetic strategies
- Chemical modification
- DDS, drug delivery systems
- ECM, extracellular matrix
- ENA, ethylene-bridged nucleic acid
- ESC, enhanced stabilization chemistry
- EVA, ethylene vinyl acetate
- Fc/HSA fusion
- FcRn, Fc receptor
- GLP-1, glucagon like peptide-1
- GS, glycine–serine
- HA, hyaluronic acid
- HES, hydroxy-ethyl-starch
- HP, hypoparathyroidism
- HSA, human serum albumin
- Hydrogels
- ISFI, in situ forming implants
- IgG, immunoglobulin G
- Implantable systems
- LAFs, long-acting formulations
- LAPFs, long-acting parenteral formulations
- LNA, locked nucleic acid
- Long-acting
- MNs, microneedles
- Microneedles
- NDS, nanochannel delivery system
- NPs, nanoparticles
- Nanocrystal suspensions
- OA, osteoarthritis
- PCPP-SA, poly(1,3-bis(carboxyphenoxy)propane-co-sebacic-acid)
- PEG, polyethylene glycol
- PM, platelet membrane
- PMPC, poly(2-methyacryloyloxyethyl phosphorylcholine)
- PNAs, peptide nucleic acids
- PS, phase separation
- PSA, polysialic acid
- PTH, parathyroid hormone
- PVA, polyvinyl alcohol
- RBCs, red blood cells
- RES, reticuloendothelial system
- RNAi, RNA interference
- SAR, structure‒activity relationship
- SCID, severe combined immunodeficiency
- SE, solvent extraction
- STC, standard template chemistry
- TNFR2, tumor necrosis factor receptor 2
- hGH, human growth hormone
- im, intramuscular
- iv, intravenous
- mPEG, methoxypolyethylene glycol
- sc, subcutaneous
Collapse
Affiliation(s)
- Yujie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiangyu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
41
|
Abstract
Drug delivery systems have shown tremendous promise to improve the diagnostic and therapeutic effects of drugs due to their special property. Targeting tissue damage, tumors, or drugs with limited toxicity at the site of infection is the goal of successful pharmaceuticals. Targeted drug delivery has become significantly important in enhancing the pharmaceutical effects of drugs and reducing their side effects of therapeutics in the treatment of various disease conditions. Unfortunately, clinical translation of these targeted drug delivery system mechanisms faces many challenges. At present, only a few targeted drug delivery systems can achieve high targeting efficiency after intravenous injection, even though numerous surface markers and targeting approaches have been developed. Thus, cell-mediated drug-delivery targeting systems have received considerable attention for their enhanced therapeutic specificity and efficacy in the treatment of the disease. This review highlights the recent advances in the design of the different types of cells that have been explored for cell-mediated drug delivery and targeting mechanisms. A better understanding of cell biology orientation and a new generation of delivery strategies that utilize these endogenous approaches are expected to provide better solutions for specific site delivery and further facilitate clinical translation.
Collapse
Affiliation(s)
- Hongli Yu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Zhihong Yang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Fei Li
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lisa Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Yong Sun
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| |
Collapse
|
42
|
Thomsen T, Reissmann R, Kaba E, Engelhardt B, Klok HA. Covalent and Noncovalent Conjugation of Degradable Polymer Nanoparticles to T Lymphocytes. Biomacromolecules 2021; 22:3416-3430. [PMID: 34170107 DOI: 10.1021/acs.biomac.1c00488] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cells are attractive as carriers that can help to enhance control over the biodistribution of polymer nanomedicines. One strategy to use cells as carriers is based on the cell surface immobilization of the nanoparticle cargo. While a range of strategies can be used to immobilize nanoparticles on cell surfaces, only limited effort has been made to investigate the effect of these surface modification chemistries on cell viability and functional properties. This study has explored seven different approaches for the immobilization of poly(lactic acid) (PLA) nanoparticles on the surface of two different T lymphocyte cell lines. The cell lines used were human Jurkat T cells and CD4+ TEM cells. The latter cells possess blood-brain barrier (BBB) migratory properties and are attractive for the development of cell-based delivery systems to the central nervous system (CNS). PLA nanoparticles were immobilized either via covalent active ester-amine, azide-alkyne cycloaddition, and thiol-maleimide coupling, or via noncovalent approaches that use lectin-carbohydrate, electrostatic, or biotin-NeutrAvidin interactions. The cell surface immobilization of the nanoparticles was monitored with flow cytometry and confocal microscopy. By tuning the initial nanoparticle/cell ratio, T cells can be decorated with up to ∼185 nanoparticles/cell as determined by confocal microscopy. The functional properties of the nanoparticle-decorated cells were assessed by evaluating their binding to ICAM-1, a key protein involved in the adhesion of CD4+ TEM cells to the BBB endothelium, as well as in a two-chamber model in vitro BBB migration assay. It was found that the migratory behavior of CD4+ TEM cells carrying carboxylic acid-, biotin-, or Wheat germ agglutinin (WGA)-functionalized nanoparticles was not affected by the presence of the nanoparticle payload. In contrast, however, for cells decorated with maleimide-functionalized nanoparticles, a reduction in the number of migratory cells compared to the nonmodified control cells was observed. Investigating and understanding the impact of nanoparticle-cell surface conjugation chemistries on the viability and properties of cells is important to further improve the design of cell-based nanoparticle delivery systems. The results of this study present a first step in this direction and provide first guidelines for the surface modification of T cells, in particular in view of their possible use for drug delivery to the CNS.
Collapse
Affiliation(s)
- Tanja Thomsen
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Regina Reissmann
- University of Bern, Theodor Kocher Institute,Freiestrasse 1, CH-3000 Bern, Switzerland
| | - Elisa Kaba
- University of Bern, Theodor Kocher Institute,Freiestrasse 1, CH-3000 Bern, Switzerland
| | - Britta Engelhardt
- University of Bern, Theodor Kocher Institute,Freiestrasse 1, CH-3000 Bern, Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|
43
|
Song S, Xia H, Guo M, Wang S, Zhang S, Ma P, Jin Y. Role of macrophage in nanomedicine-based disease treatment. Drug Deliv 2021; 28:752-766. [PMID: 33860719 PMCID: PMC8079019 DOI: 10.1080/10717544.2021.1909175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Macrophages are a major component of the immunoresponse. Diversity and plasticity are two of the hallmarks of macrophages, which allow them to act as proinflammatory, anti-inflammatory, and homeostatic agents. Research has found that cancer and many inflammatory or autoimmune disorders are correlated with activation and tissue infiltration of macrophages. Recent developments in macrophage nanomedicine-based disease treatment are proving to be timely owing to the increasing inadequacy of traditional treatment. Here, we review the role of macrophages in nanomedicine-based disease treatment. First, we present a brief background on macrophages and nanomedicine. Then, we delve into applications of macrophages as a target for disease treatment and delivery systems and summarize the applications of macrophage-derived extracellular vesicles. Finally, we provide an outlook on the clinical utility of macrophages in nanomedicine-based disease treatment.
Collapse
Affiliation(s)
- Siwei Song
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujing Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Liang T, Zhang R, Liu X, Ding Q, Wu S, Li C, Lin Y, Ye Y, Zhong Z, Zhou M. Recent Advances in Macrophage-Mediated Drug Delivery Systems. Int J Nanomedicine 2021; 16:2703-2714. [PMID: 33854316 PMCID: PMC8039204 DOI: 10.2147/ijn.s298159] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/14/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages have been extensively used in the development of drug delivery systems, as they can prolong the circulation and release of drugs, extend their half-life, increase their stability and targeting ability, and reduce immunogenicity. Moreover, they have good biocompatibility and degradability and offer abundant surface receptors for targeted delivery of a wide variety of drugs. Macrophage-mediated drug delivery systems can be prepared by loading drugs or drug-loaded nanoparticles into macrophages, macrophage membranes or macrophage-derived vesicles. Although such systems can be used to treat inflammation, cancer, HIV infection and other diseases, they require further research and optimization since they have been assembled from diverse sources and therefore can have quite different physical and chemical properties. Moreover, potential cell-drug interactions can limit their application, and the biological activity of membrane proteins might be lost during membrane extraction and storage. In this review, we summarize the recent advances in this field and discuss the preparation of macrophage-mediated drug delivery systems, their advantages over other delivery systems, their potential applications and future lines of research.
Collapse
Affiliation(s)
- Tiantian Liang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Rongtao Zhang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Xianbin Liu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Qian Ding
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Siqiong Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Yan Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Yun Ye
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China
| |
Collapse
|
45
|
Jahromi LP, Shahbazi M, Maleki A, Azadi A, Santos HA. Chemically Engineered Immune Cell-Derived Microrobots and Biomimetic Nanoparticles: Emerging Biodiagnostic and Therapeutic Tools. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002499. [PMID: 33898169 PMCID: PMC8061401 DOI: 10.1002/advs.202002499] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/26/2020] [Indexed: 05/16/2023]
Abstract
Over the past decades, considerable attention has been dedicated to the exploitation of diverse immune cells as therapeutic and/or diagnostic cell-based microrobots for hard-to-treat disorders. To date, a plethora of therapeutics based on alive immune cells, surface-engineered immune cells, immunocytes' cell membranes, leukocyte-derived extracellular vesicles or exosomes, and artificial immune cells have been investigated and a few have been introduced into the market. These systems take advantage of the unique characteristics and functions of immune cells, including their presence in circulating blood and various tissues, complex crosstalk properties, high affinity to different self and foreign markers, unique potential of their on-demand navigation and activity, production of a variety of chemokines/cytokines, as well as being cytotoxic in particular conditions. Here, the latest progress in the development of engineered therapeutics and diagnostics inspired by immune cells to ameliorate cancer, inflammatory conditions, autoimmune diseases, neurodegenerative disorders, cardiovascular complications, and infectious diseases is reviewed, and finally, the perspective for their clinical application is delineated.
Collapse
Affiliation(s)
- Leila Pourtalebi Jahromi
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShiraz71468‐64685Iran
- Present address:
Helmholtz Institute for Pharmaceutical Research SaarlandHelmholtz Centre for Infection ResearchBiogenic Nanotherapeutics GroupCampus E8.1Saarbrücken66123Germany
| | - Mohammad‐Ali Shahbazi
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC)Zanjan University of Medical SciencesZanjan45139‐56184Iran
| | - Aziz Maleki
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC)Zanjan University of Medical SciencesZanjan45139‐56184Iran
| | - Amir Azadi
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShiraz71468‐64685Iran
- Department of PharmaceuticsSchool of PharmacyShiraz University of Medical SciencesShiraz71468‐64685Iran
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFI‐00014Finland
| |
Collapse
|
46
|
Su Y, Zhang T, Huang T, Gao J. Current advances and challenges of mesenchymal stem cells-based drug delivery system and their improvements. Int J Pharm 2021; 600:120477. [PMID: 33737099 DOI: 10.1016/j.ijpharm.2021.120477] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as a promising living carrier for targeted drug delivery. A wealth of literature has shown evidence for great advances in MSCs-based drug delivery system (MSCs-DDS) in the treatment of various diseases. Nevertheless, as this field of study rapidly advances, several challenges associated with this delivery strategy have arisen, mainly due to the inherent limitations of MSCs. To this end, several novel technologies are being developed in parallel to improve the efficiency or safety of this system. In this review, we introduce recent advances and summarize the present challenges of MSCs-DDS. We also highlight some potential technologies to improve MSCs-DDS, including nanotechnology, genome engineering technology, and biomimetic technology. Finally, prospects for application of artificially improved MSCs-DDS are addressed. The technologies summarized in this review provide a general guideline for the improvement of MSCs-DDS.
Collapse
Affiliation(s)
- Yuanqin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
47
|
Fan XM, Shen JJ, Xu YY, Gao J, Zhang YW. Metabolic integration of azide functionalized glycan on Escherichia coli cell surface for specific covalent immobilization onto magnetic nanoparticles with click chemistry. BIORESOURCE TECHNOLOGY 2021; 324:124689. [PMID: 33450627 DOI: 10.1016/j.biortech.2021.124689] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 06/12/2023]
Abstract
A method for specific immobilization of whole-cell with covalent bonds was developed through a click reaction between alkyne and azide groups. In this approach, magnetic nanoparticle Fe3O4@SiO2-NH2-alkyne was synthesized with Fe3O4 core preparation, SiO2 coating, and alkyne functionalization on the surface. The azides were successfully integrated onto the cell surface of the recombinant E. coli harboring glycerol dehydrogenase, which was employed as the model cell. The highest immobilization yield of 83% and activity recovery of 94% were obtained under the conditions of 0.67 mg mg-1 cell-support ratio, pH 6.0, temperature 45 °C, and 20 mM Cu2+ concentration. The immobilized cell showed good reusability, which remained over 50% of initial activity after 10 cycles of utilization. Its activity was 9.7-fold higher than that of the free cell at the condition of pH 8.0 and each optimal temperature. Furthermore, the immobilized cell showed significantly higher activity, operational stability, and reusability.
Collapse
Affiliation(s)
- Xiao-Man Fan
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Jia-Jia Shen
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Yuan-Yuan Xu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Jian Gao
- College of Petroleum and Chemical Engineering, Beibu Gulf University, 535100 Qinzhou, People's Republic of China
| | - Ye-Wang Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, People's Republic of China; College of Petroleum and Chemical Engineering, Beibu Gulf University, 535100 Qinzhou, People's Republic of China.
| |
Collapse
|
48
|
Ayer M, Burri O, Guiet R, Seitz A, Kaba E, Engelhardt B, Klok HA. Biotin-NeutrAvidin Mediated Immobilization of Polymer Micro- and Nanoparticles on T Lymphocytes. Bioconjug Chem 2021; 32:541-552. [PMID: 33621057 DOI: 10.1021/acs.bioconjchem.1c00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cells are powerful carriers that can help to improve the delivery of nanomedicines. One approach to use cells as carriers is to immobilize the nanoparticulate cargo on the cell surface. While a plethora of chemical conjugation strategies are available to bind nanoparticles to cell surfaces, only relatively little is known about the effects of particle size and cell type on the surface immobilization of nanoparticles. This study investigates the biotin-NeutrAvidin mediated immobilization of model polymer nanoparticles with sizes ranging from 40 nm to 1 μm on two different T cell lines, viz., human Jurkat cells as well as mouse SJL/PLP7 T cells, which are of potential interest for drug delivery across the blood-brain barrier. The nanoparticle cell surface immobilization and the particle surface concentration and distribution were analyzed by flow cytometry and confocal microscopy. The functional properties of nanoparticle-modified SJL/PLP7 T cells were assessed in an ICAM-1 binding assay as well as in a two-chamber setup in which the migration of the particle-modified T cells across an in vitro model of the blood-brain barrier was studied. The results of these experiments highlight the effects of particle size and cell line on the surface immobilization of nanoparticles on living cells.
Collapse
Affiliation(s)
- Maxime Ayer
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Olivier Burri
- École Polytechnique Fédérale de Lausanne (EPFL), Faculté des Sciences de la Vie, Bioimaging and Optics Platform, Bâtiment AI, Station 15, CH-1015 Lausanne, Switzerland
| | - Romain Guiet
- École Polytechnique Fédérale de Lausanne (EPFL), Faculté des Sciences de la Vie, Bioimaging and Optics Platform, Bâtiment AI, Station 15, CH-1015 Lausanne, Switzerland
| | - Arne Seitz
- École Polytechnique Fédérale de Lausanne (EPFL), Faculté des Sciences de la Vie, Bioimaging and Optics Platform, Bâtiment AI, Station 15, CH-1015 Lausanne, Switzerland
| | - Elisa Kaba
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|
49
|
Sang X, Wang Y, Xue Z, Qi D, Fan G, Tian F, Zhu Y, Yang J. Macrophage-Targeted Lung Delivery of Dexamethasone Improves Pulmonary Fibrosis Therapy via Regulating the Immune Microenvironment. Front Immunol 2021; 12:613907. [PMID: 33679754 PMCID: PMC7935565 DOI: 10.3389/fimmu.2021.613907] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is serious chronic lung disease with limited therapeutic approaches. Inflammation and immune disorders are considered as the main factors in the initiation and development of pulmonary fibrosis. Inspired by the key roles of macrophages during the processes of inflammation and immune disorders, here, we report a new method for direct drug delivery into the in-situ fibrotic tissue sites in vitro and in vivo. First, liposomes containing dexamethasone (Dex-L) are prepared and designed to entry into the macrophages in the early hours, forming the macrophages loaded Dex-L delivery system (Dex-L-MV). Chemokine and cytokine factors such as IL-6, IL-10, Arg-1 are measured to show the effect of Dex-L to the various subtypes of macrophages. Next, we mimic the inflammatory and anti-inflammatory microenvironment by co-culture of polarized/inactive macrophage and fibroblast cells to show the acute inflammation response of Dex-L-MV. Further, we confirm the targeted delivery of Dex-L-MV into the inflammatory sites in vivo, and surprisingly found that injected macrophage containing Dex can reduce the level of macrophage infiltration and expression of the markers of collagen deposition during the fibrotic stage, while causing little systematic toxicity. These data demonstrated the suitability and immune regulation effect of Dex-L-MV for the anti-pulmonary process. It is envisaged that these findings are a step forward toward endogenous immune targeting systems as a tool for clinical drug delivery.
Collapse
Affiliation(s)
- Xiaoqing Sang
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanyuan Wang
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhifeng Xue
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dawei Qi
- Medcity Research Laboratory, University of Turku, Turku, Finland
| | - Guanwei Fan
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of Traditional Chinese Medicine Prescription and Syndrome, Tianjin, China
| | - Fei Tian
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Traditional Chinese Medicine Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jian Yang
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
50
|
Thomsen T, Klok HA. Chemical Cell Surface Modification and Analysis of Nanoparticle-Modified Living Cells. ACS APPLIED BIO MATERIALS 2021; 4:2293-2306. [DOI: 10.1021/acsabm.0c01619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Tanja Thomsen
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|