1
|
Chen Q, Zheng Y, Jiang X, Wang Y, Chen Z, Wu D. Nature's carriers: leveraging extracellular vesicles for targeted drug delivery. Drug Deliv 2024; 31:2361165. [PMID: 38832506 DOI: 10.1080/10717544.2024.2361165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
With the rapid development of drug delivery systems, extracellular vesicles (EVs) have emerged as promising stars for improving targeting abilities and realizing effective delivery. Numerous studies have shown when compared to conventional strategies in targeted drug delivery (TDD), EVs-based strategies have several distinguished advantages besides targeting, such as participating in cell-to-cell communications and immune response, showing high biocompatibility and stability, penetrating through biological barriers, etc. In this review, we mainly focus on the mass production of EVs including the challenges and strategies for scaling up EVs production in a cost-effective and reproducible manner, the loading and active targeting methods, and examples of EVs as vehicles for TDD in consideration of potential safety and regulatory issues associated. We also conclude and discuss the rigor and reproducibility of EVs production, the current research status of the application of EVs-based strategies to targeted drug delivery, clinical conversion prospects, and existing chances and challenges.
Collapse
Affiliation(s)
- Qi Chen
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, P. R. China
| | - Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuhong Jiang
- Epilepsy Center, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, PR China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Epilepsy Center, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Sun S, Han R, Sun Y, Chen W, Zhao L, Guan X, Zhang W. A minimalist cancer cell membrane-shielded biomimetic nanoparticle for nasopharyngeal carcinoma active-targeting therapy. Colloids Surf B Biointerfaces 2024; 238:113909. [PMID: 38599076 DOI: 10.1016/j.colsurfb.2024.113909] [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: 02/20/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a common head and neck malignancy, which is characterized by high incidence and aggression with poor diagnosis and limited therapeutic opportunity. The innovative strategy for achieving precise NPC active-targeting drug delivery has emerged as a prominent focus in clinical research. Here, a minimalist cancer cell membrane (CCM) shielded biomimetic nanoparticle (NP) was designed for NPC active-targeting therapy. Chemotherapeutant model drug doxorubicin (DOX) was loaded in polyamidoamine (PAMAM) dendrimer. The PAMAM/DOX (PD) NP was further shielded by human CNE-2 NPC CCM. Characterization results verified that the biomimetic PAMAM/DOX@CCM (abbreviated as PDC) NPs had satisfactory physical properties with high DOX-loading and excellent stability. Cell experiments demonstrated that the CNE-2 membrane-cloaked PDC NPs presented powerful cellular uptake in the sourcing cells by homologous targeting and adhesive interaction. Further in vivo results confirmed that this biomimetic nanoplatform had extended circulation and remarkable tumor-targeting capability, and the PDC NPs effectively suppressed the progression of CNE-2 tumors by systemic administration. This CCM-shielded biomimetic NP displayed a minimalist paradigm nanoplatform for precise NPC therapy, and the strategy of CCM-shielded biomimetic drug delivery system (DDS) has great potential for extensive cancer active-targeting therapy.
Collapse
Affiliation(s)
- Shuo Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Rongrong Han
- Department of Otolaryngology, Weifang People's Hospital, Weifang 261000, China
| | - Yanju Sun
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Wenqiang Chen
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Limin Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Xiuwen Guan
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| | - Weifen Zhang
- College of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
3
|
Wang C, Liu H, Lin H, Zhong R, Li H, Liu J, Luo X, Tian M. Effect of zwitterionic sulfobetaine incorporation on blood behaviours, phagocytosis, and in vivo biodistribution of pH-responsive micelles with positive charges. J Mater Chem B 2024; 12:1652-1666. [PMID: 38275277 DOI: 10.1039/d3tb02477f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
pH-responsive micelles with positive charges are challenged by their significant effect on the cells/proteins and compromise their final fate due to electrostatic interactions. As one of the promising strategies, zwitterion incorporation in micelles has attracted considerable attention and displayed improved protein adsorption and blood circulation performances. However, previous reports in this field have been mostly limited in hemolysis for studying blood behaviour and lack a comprehensive understanding of their interactions with blood components. Herein, we present a prelimilary study on the effect of zwitterionic sulfobetaine incorporation on blood behaviour, phagocytosis, and in vivo biodistribution of pH-responsive micelles with positive charges. Amphiphilic triblock copolymers, namely poly(ε-caprolactone)-b-poly(N,N-diethylaminoethyl methacrylate)-(N-(3-sulfopropyl-N-methacryloxyethy-N,N-diethylammonium betaine)) (PCL-PDEAPSx, x = 2, 6, 10), containing different numbers of sulfobetaine groups were synthesized through four steps to prepare the pH-responsive micelles with positive charges. The effect of the sulfobetaine incorporation displayed different profiles, e.g., the micelles had no effect on RBC aggregation, thrombin time (TT), and platelet aggregation, while the cytotoxicity, hemolysis, RBC deformability, activated partial thromboplastin time (APTT), prothrombin time (PT), platelet activation, protein (albumin, fibrinogen, plasma) adsorption, phagocytosis, and in vivo biodistribution decreased with the increase in the sulfobetaine number, in which the transition mainly occurred at a sulfobetaine/tertiary amine group ratio of 3/7-1/1 compared to that of the mPEG control. In addition, the micelles displayed a strong inhibitory effect on the intrinsic coagulation pathway, which was associated with a significant decrease in the coagulation factor activity. Based on these findings, the related mechanism is discussed and proposed, which can aid the rational design of pH-responsive micelles for improved therapeutics.
Collapse
Affiliation(s)
- Chengwei Wang
- Division of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Hao Liu
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China.
| | - Hu Lin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Rui Zhong
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, 610052, P. R. China
| | - Hao Li
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China.
| | - Jiaxin Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, 610052, P. R. China
| | - Xianglin Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Meng Tian
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China.
| |
Collapse
|
4
|
Ouyang X, Liu Y, Zheng K, Pang Z, Peng S. Recent advances in zwitterionic nanoscale drug delivery systems to overcome biological barriers. Asian J Pharm Sci 2024; 19:100883. [PMID: 38357524 PMCID: PMC10861844 DOI: 10.1016/j.ajps.2023.100883] [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: 02/09/2023] [Revised: 05/28/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024] Open
Abstract
Nanoscale drug delivery systems (nDDS) have been employed widely in enhancing the therapeutic efficacy of drugs against diseases with reduced side effects. Although several nDDS have been successfully approved for clinical use up to now, biological barriers between the administration site and the target site hinder the wider clinical adoption of nDDS in disease treatment. Polyethylene glycol (PEG)-modification (or PEGylation) has been regarded as the gold standard for stabilising nDDS in complex biological environment. However, the accelerated blood clearance (ABC) of PEGylated nDDS after repeated injections becomes great challenges for their clinical applications. Zwitterionic polymer, a novel family of anti-fouling materials, have evolved as an alternative to PEG due to their super-hydrophilicity and biocompatibility. Zwitterionic nDDS could avoid the generation of ABC phenomenon and exhibit longer blood circulation time than the PEGylated analogues. More impressively, zwitterionic nDDS have recently been shown to overcome multiple biological barriers such as nonspecific organ distribution, pressure gradients, impermeable cell membranes and lysosomal degradation without the need of any complex chemical modifications. The realization of overcoming multiple biological barriers by zwitterionic nDDS may simplify the current overly complex design of nDDS, which could facilitate their better clinical translation. Herein, we summarise the recent progress of zwitterionic nDDS at overcoming various biological barriers and analyse their underlying mechanisms. Finally, prospects and challenges are introduced to guide the rational design of zwitterionic nDDS for disease treatment.
Collapse
Affiliation(s)
- Xumei Ouyang
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China
| | - Yu Liu
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China
| | - Ke Zheng
- School of Materials Science and Engineering, Dongguan University of Technology, Dongguan 523808, China
| | - Zhiqing Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China
| |
Collapse
|
5
|
Banat H, Csóka I, Paróczai D, Burian K, Farkas Á, Ambrus R. A Novel Combined Dry Powder Inhaler Comprising Nanosized Ketoprofen-Embedded Mannitol-Coated Microparticles for Pulmonary Inflammations: Development, In Vitro-In Silico Characterization, and Cell Line Evaluation. Pharmaceuticals (Basel) 2024; 17:75. [PMID: 38256908 PMCID: PMC10818896 DOI: 10.3390/ph17010075] [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: 12/19/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Pulmonary inflammations such as chronic obstructive pulmonary disease and cystic fibrosis are widespread and can be fatal, especially when they are characterized by abnormal mucus accumulation. Inhaled corticosteroids are commonly used for lung inflammations despite their considerable side effects. By utilizing particle engineering techniques, a combined dry powder inhaler (DPI) comprising nanosized ketoprofen-embedded mannitol-coated microparticles was developed. A nanoembedded microparticle system means a novel advance in pulmonary delivery by enhancing local pulmonary deposition while avoiding clearance mechanisms. Ketoprofen, a poorly water-soluble anti-inflammatory drug, was dispersed in the stabilizer solution and then homogenized by ultraturrax. Following this, a ketoprofen-containing nanosuspension was produced by wet-media milling. Furthermore, co-spray drying was conducted with L-leucine (dispersity enhancer) and mannitol (coating and mucuactive agent). Particle size, morphology, dissolution, permeation, viscosity, in vitro and in silico deposition, cytotoxicity, and anti-inflammatory effect were investigated. The particle size of the ketoprofen-containing nanosuspension was ~230 nm. SEM images of the spray-dried powder displayed wrinkled, coated, and nearly spherical particles with a final size of ~2 µm (nano-in-micro), which is optimal for pulmonary delivery. The mannitol-containing samples decreased the viscosity of 10% mucin solution. The results of the mass median aerodynamic diameter (2.4-4.5 µm), fine particle fraction (56-71%), permeation (five-fold enhancement), and dissolution (80% release in 5 min) confirmed that the system is ideal for local inhalation. All samples showed a significant anti-inflammatory effect and decreased IL-6 on the LPS-treated U937 cell line with low cytotoxicity. Hence, developing an innovative combined DPI comprising ketoprofen and mannitol by employing a nano-in-micro approach is a potential treatment for lung inflammations.
Collapse
Affiliation(s)
- Heba Banat
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u.6, 6720 Szeged, Hungary; (H.B.); (I.C.)
| | - Ildikó Csóka
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u.6, 6720 Szeged, Hungary; (H.B.); (I.C.)
| | - Dóra Paróczai
- Department of Medical Microbiology, Faculty of Medicine, University of Szeged, Dóm Square 10, 6720 Szeged, Hungary; (D.P.); (K.B.)
| | - Katalin Burian
- Department of Medical Microbiology, Faculty of Medicine, University of Szeged, Dóm Square 10, 6720 Szeged, Hungary; (D.P.); (K.B.)
| | - Árpád Farkas
- Centre for Energy Research, Hungarian Academy of Sciences, 1121 Budapest, Hungary;
| | - Rita Ambrus
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u.6, 6720 Szeged, Hungary; (H.B.); (I.C.)
| |
Collapse
|
6
|
Song Q, Yang J, Wu X, Li Y, Zhao H, Feng Q, Zhang Z, Zhang Y, Wang L. A multifunctional integrated biomimetic spore nanoplatform for successively overcoming oral biological barriers. J Nanobiotechnology 2023; 21:302. [PMID: 37641137 PMCID: PMC10463901 DOI: 10.1186/s12951-023-01995-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/10/2023] [Indexed: 08/31/2023] Open
Abstract
The biological barriers have seriously restricted the efficacious responses of oral delivery system in diseases treatment. Utilizing a carrier based on the single construction means is hard to overcome these obstacles simultaneously because the complex gastrointestinal tract environment requires carrier to have different or even contradictory properties. Interestingly, spore capsid (SC) integrates many unique biological characteristics, such as high resistance, good stability etc. This fact offers a boundless source of inspiration for the construction of multi-functional oral nanoplatform based on SC without further modification. Herein, we develop a type of biomimetic spore nanoplatform (SC@DS NPs) to successively overcome oral biological barriers. Firstly, doxorubicin (DOX) and sorafenib (SOR) are self-assembled to form carrier-free nanoparticles (DS NPs). Subsequently, SC is effectively separated from probiotic spores and served as a functional vehicle for delivering DS NPs. As expect, SC@DS NPs can efficaciously pass through the rugged stomach environment after oral administration and further be transported to the intestine. Surprisingly, we find that SC@DS NPs exhibit a significant improvement in the aspects of mucus penetration and transepithelial transport, which is related to the protein species of SC. This study demonstrates that SC@DS NPs can efficiently overcome multiple biological barriers and improve the therapeutic effect.
Collapse
Affiliation(s)
- Qingling Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China
| | - Junfei Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
| | - Xiaocui Wu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
| | - Yao Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
| | - Hongjuan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China
| | - Qianhua Feng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China.
| | - Yun Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China.
| | - Lei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China.
| |
Collapse
|
7
|
Chen Y, Tao H, Chen R, Pan Y, Wang J, Gao R, Chen J, Yang J. Biomimetic Nanoparticles Loaded with Ulinastatin for the Targeted Treatment of Acute Pancreatitis. Mol Pharm 2023; 20:4108-4119. [PMID: 37349264 DOI: 10.1021/acs.molpharmaceut.3c00238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Ulinastatin is commonly used in the clinic to treat acute pancreatitis (AP), but its therapeutic effect was limited by the presence of the blood-pancreas barrier (BPB) and low specificity. Here, we prepared a macrophage biomimetic nanoparticle (MU) that delivered ulinastatin to address the above issues. Macrophage membrane was used as a shell for a mixture of PEG-PLGA and ulinastatin. It was found that MU showed good stability and biocompatibility in vitro and in vivo. According to in vivo fluorescence imaging, MU displayed a great inflammation targeting effect both in a subcutaneous inflammation model and in situ pancreatitis mouse model, which was ascribed to the presence of adhesion proteins. In vitro and in vivo results demonstrated that MU have a superior AP treatment effect by inhibiting pro-inflammatory factors and keeping cells viability. It was suggested the MU could provide a new strategy for targeted AP treatment.
Collapse
Affiliation(s)
- Yunlong Chen
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Haisu Tao
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832000, China
| | - Rui Chen
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Biliary Surgical Department of West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yingying Pan
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Junfeng Wang
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Rongkang Gao
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jingqin Chen
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jian Yang
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
8
|
Abstract
Intoxication is one of the most common causes of accidental death globally. Although some antidotes capable of neutralizing the toxicity of certain xenobiotics have become well established, the current reality is that clinicians primarily rely on nonspecific extracorporeal techniques to remove toxins. Nano-intervention strategies in which nanoantidotes neutralize toxicity in situ via physical interaction, chemical bonding, or biomimetic clearance have begun to show clinical potential. However, most nanoantidotes remain in the proof-of-concept stage, and the difficulty of constructing clinical relevance models and the unclear pharmacokinetics of nanoantidotes hinder their translation to clinic. This Concept reviews the detoxification mechanisms of polymer nanoantidotes and predicts the opportunities and challenges associated with their clinical application.
Collapse
Affiliation(s)
- Jiazhen Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| | - Hongjie Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| | - Haoyang Zou
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| |
Collapse
|
9
|
Feng C, Tan P, Nie G, Zhu M. Biomimetic and bioinspired nano-platforms for cancer vaccine development. EXPLORATION (BEIJING, CHINA) 2023; 3:20210263. [PMID: 37933383 PMCID: PMC10624393 DOI: 10.1002/exp.20210263] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2023]
Abstract
The advent of immunotherapy has revolutionized the treating modalities of cancer. Cancer vaccine, aiming to harness the host immune system to induce a tumor-specific killing effect, holds great promises for its broad patient coverage, high safety, and combination potentials. Despite promising, the clinical translation of cancer vaccines faces obstacles including the lack of potency, limited options of tumor antigens and adjuvants, and immunosuppressive tumor microenvironment. Biomimetic and bioinspired nanotechnology provides new impetus for the designing concepts of cancer vaccines. Through mimicking the stealth coating, pathogen recognition pattern, tissue tropism of pathogen, and other irreplaceable properties from nature, biomimetic and bioinspired cancer vaccines could gain functions such as longstanding, targeting, self-adjuvanting, and on-demand cargo release. The specific behavior and endogenous molecules of each type of living entity (cell or microorganism) offer unique features to cancer vaccines to address specific needs for immunotherapy. In this review, the strategies inspired by eukaryotic cells, bacteria, and viruses will be overviewed for advancing cancer vaccine development. Our insights into the future cancer vaccine development will be shared at the end for expediting the clinical translation.
Collapse
Affiliation(s)
- Chenchao Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Peng Tan
- Klarman Cell ObservatoryBroad Institute of MIT and HarvardCambridgeUSA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
- GBA Research Innovation Institute for NanotechnologyGuangzhouChina
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
| |
Collapse
|
10
|
Song W, Jia P, Ren Y, Xue J, Zhou B, Xu X, Shan Y, Deng J, Zhou Q. Engineering white blood cell membrane-camouflaged nanocarriers for inflammation-related therapeutics. Bioact Mater 2023; 23:80-100. [DOI: 10.1016/j.bioactmat.2022.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/11/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
|
11
|
Chen BQ, Zhao Y, Zhang Y, Pan YJ, Xia HY, Kankala RK, Wang SB, Liu G, Chen AZ. Immune-regulating camouflaged nanoplatforms: A promising strategy to improve cancer nano-immunotherapy. Bioact Mater 2023; 21:1-19. [PMID: 36017071 PMCID: PMC9382433 DOI: 10.1016/j.bioactmat.2022.07.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/11/2022] [Accepted: 07/24/2022] [Indexed: 02/06/2023] Open
Abstract
Although nano-immunotherapy has advanced dramatically in recent times, there remain two significant hurdles related to immune systems in cancer treatment, such as (namely) inevitable immune elimination of nanoplatforms and severely immunosuppressive microenvironment with low immunogenicity, hampering the performance of nanomedicines. To address these issues, several immune-regulating camouflaged nanocomposites have emerged as prevailing strategies due to their unique characteristics and specific functionalities. In this review, we emphasize the composition, performances, and mechanisms of various immune-regulating camouflaged nanoplatforms, including polymer-coated, cell membrane-camouflaged, and exosome-based nanoplatforms to evade the immune clearance of nanoplatforms or upregulate the immune function against the tumor. Further, we discuss the applications of these immune-regulating camouflaged nanoplatforms in directly boosting cancer immunotherapy and some immunogenic cell death-inducing immunotherapeutic modalities, such as chemotherapy, photothermal therapy, and reactive oxygen species-mediated immunotherapies, highlighting the current progress and recent advancements. Finally, we conclude the article with interesting perspectives, suggesting future tendencies of these innovative camouflaged constructs towards their translation pipeline.
Collapse
Affiliation(s)
- Biao-Qi Chen
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Yi Zhao
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, PR China
| | - Yu-Jing Pan
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, PR China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China
| |
Collapse
|
12
|
4T1 cell membrane-derived biodegradable nanosystem for comprehensive interruption of cancer cell metabolism. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
13
|
Jampilek J, Kralova K. Insights into Lipid-Based Delivery Nanosystems of Protein-Tyrosine Kinase Inhibitors for Cancer Therapy. Pharmaceutics 2022; 14:2706. [PMID: 36559200 PMCID: PMC9783038 DOI: 10.3390/pharmaceutics14122706] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022] Open
Abstract
According to the WHO, cancer caused almost 10 million deaths worldwide in 2020, i.e., almost one in six deaths. Among the most common are breast, lung, colon and rectal and prostate cancers. Although the diagnosis is more perfect and spectrum of available drugs is large, there is a clear trend of an increase in cancer that ends fatally. A major advance in treatment was the introduction of gentler antineoplastics for targeted therapy-tyrosine kinase inhibitors (TKIs). Although they have undoubtedly revolutionized oncology and hematology, they have significant side effects and limited efficacy. In addition to the design of new TKIs with improved pharmacokinetic and safety profiles, and being more resistant to the development of drug resistance, high expectations are placed on the reformulation of TKIs into various drug delivery lipid-based nanosystems. This review provides an insight into the history of chemotherapy, a brief overview of the development of TKIs for the treatment of cancer and their mechanism of action and summarizes the results of the applications of self-nanoemulsifying drug delivery systems, nanoemulsions, liposomes, solid lipid nanoparticles, lipid-polymer hybrid nanoparticles and nanostructured lipid carriers used as drug delivery systems of TKIs obtained in vitro and in vivo.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| | - Katarina Kralova
- Institute of Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
14
|
Dessale M, Mengistu G, Mengist HM. Nanotechnology: A Promising Approach for Cancer Diagnosis, Therapeutics and Theragnosis. Int J Nanomedicine 2022; 17:3735-3749. [PMID: 36051353 PMCID: PMC9427008 DOI: 10.2147/ijn.s378074] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/22/2022] [Indexed: 01/10/2023] Open
Abstract
Cancer remains the most devastating disease and the major cause of mortality worldwide. Although early diagnosis and treatment are the key approach in fighting against cancer, the available conventional diagnostic and therapeutic methods are not efficient. Besides, ineffective cancer cell selectivity and toxicity of traditional chemotherapy remain the most significant challenge. These limitations entail the need for the development of both safe and effective cancer diagnosis and treatment options. Due to its robust application, nanotechnology could be a promising method for in-vivo imaging and detection of cancer cells and cancer biomarkers. Nanotechnology could provide a quick, safe, cost-effective, and efficient method for cancer management. It also provides simultaneous diagnosis and treatment of cancer using nano-theragnostic particles that facilitate early detection and selective destruction of cancer cells. Updated and recent discussions are important for selecting the best cancer diagnosis, treatment, and management options, and new insights on designing effective protocols are utmost important. This review discusses the application of nanotechnology in cancer diagnosis, therapeutics, and theragnosis and provides future perspectives in the field.
Collapse
Affiliation(s)
- Mesfin Dessale
- Department of Medical Laboratory Sciences, Debre Markos University, Debre Markos, Amhara, Ethiopia
| | - Getachew Mengistu
- Department of Medical Laboratory Sciences, Debre Markos University, Debre Markos, Amhara, Ethiopia
| | | |
Collapse
|
15
|
Zhang W, Huang X. Stem cell membrane-camouflaged targeted delivery system in tumor. Mater Today Bio 2022; 16:100377. [PMID: 35967738 PMCID: PMC9364095 DOI: 10.1016/j.mtbio.2022.100377] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023] Open
|
16
|
Hou Z, Zhou W, Guo X, Zhong R, Wang A, Li J, Cen Y, You C, Tan H, Tian M. Poly(ε-Caprolactone)-Methoxypolyethylene Glycol (PCL-MPEG)-Based Micelles for Drug-Delivery: The Effect of PCL Chain Length on Blood Components, Phagocytosis, and Biodistribution. Int J Nanomedicine 2022; 17:1613-1632. [PMID: 35411141 PMCID: PMC8994631 DOI: 10.2147/ijn.s349516] [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: 11/26/2021] [Accepted: 03/07/2022] [Indexed: 12/27/2022] Open
Abstract
Background The main challenge of polymeric micelles as drug delivery systems is that the actual delivery efficiency is not as high as expected, which is closely related with the interactions with the complex biological environments such as blood components, phagocytosis, and biodistribution. Herein, we expect to understand these concerns for the clinically relevant micelles that composed of methoxypolyethylene glycol (MPEG) with identical chain length And poly(ε-caprolactone) (PCL) with tunable chain length (PCLn-MPEG) (n=20, 30, and 40) wherein doxorubicin was encapsulated as a model drug. Methods The doxorubicin-loaded PCLn-MPEG micelles were prepared by a dialysis method and characterized by dynamic light scattering and transmission electron microscopy. The surface PEG density and chain conformation were investigated by dissipative particle dynamics simulation. The stability of the micelles was detected by nanoparticle tracking analysis. The effects of PCL chain length on the blood components, phagocytosis, and biodistribution were assayed in vitro and in vivo. Results The micelles exhibited spherical morphology with a diameter about 30nm. The PEG chain conformation from "mushroom-like" to "brush-like" was evident. The micelles have no remarkable effect on the red blood cells, blood coagulation, and platelet activation. Interestingly, the protein adsorption was affected and dependent on the chain conformation, with lowest adsorption for PCL30-MPEG, which also has the loWest phagocytosis. The stability of the micelles was in the order of PCL40-MPEG>PCL30-MPEG>PCL20-MPEG which was dependent on the PCL chain length. The micelles mainly accumulated in liver, with the order consistent with their stability, indicating that, besides the phagocytosis, the stability of the micelle plays an important role in biodistribution as well. The related mechanisms were proposed and discussed. Conclusion Manipulating the PEG/PCL ratio of the micelle is an effective approach to modulate the protein adsorption, phagocytosis, and biodistribution, which may be a prerequisite for clinical applications.
Collapse
Affiliation(s)
- Zemin Hou
- Department of Burn and Plastic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Wencheng Zhou
- Department of Burn and Plastic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xi Guo
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Rui Zhong
- Institute of Blood Transfusion, Chinese Academy of Medical Science & Peking Union Medical College, Chengdu, Sichuan, People’s Republic of China
| | - Ao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jiehua Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Ying Cen
- Department of Burn and Plastic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Chao You
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Meng Tian
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
17
|
Sun L, Lei Y, Wang Y, Liu D. Blood-based Alzheimer's disease diagnosis using fluorescent peptide nanoparticle arrays. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
18
|
Zhao J, Tan W, Zheng J, Su Y, Cui M. Aptamer Nanomaterials for Ovarian Cancer Target Theranostics. Front Bioeng Biotechnol 2022; 10:884405. [PMID: 35419352 PMCID: PMC8996158 DOI: 10.3389/fbioe.2022.884405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/14/2022] [Indexed: 12/05/2022] Open
Abstract
Ovarian cancer is among the leading causes of gynecological cancer-related mortality worldwide. Early and accurate diagnosis and an effective treatment strategy are the two primary means of improving the prognosis of patients with ovarian cancer. The development of targeted nanomaterials provides a potentially efficient strategy for ovarian cancer theranostics. Aptamer nanomaterials have emerged as promising nanoplatforms for accurate ovarian cancer diagnosis by recognizing relevant biomarkers in the serum and/or on the surface of tumor cells, as well as for effective ovarian cancer inhibition via target protein blockade on tumor cells and targeted delivery of various therapeutic agents. In this review, we summarize recent advances in aptamer nanomaterials as targeted theranostic platforms for ovarian cancer and discusses the challenges and opportunities for their clinical application. The information presented in this review represents a valuable reference for creation of a new generation of aptamer nanomaterials for use in the precise detection and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Gynecology and Obstetrics, the Second Hospital of Jilin University, Changchun, China
| | - Wenxi Tan
- Department of Gynecology and Obstetrics, the Second Hospital of Jilin University, Changchun, China
| | - Jingying Zheng
- Department of Gynecology and Obstetrics, the Second Hospital of Jilin University, Changchun, China
| | - Yuanzhen Su
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Manhua Cui
- Department of Gynecology and Obstetrics, the Second Hospital of Jilin University, Changchun, China,*Correspondence: Manhua Cui,
| |
Collapse
|
19
|
Xu S, Liu B, Fan J, Xue C, Lu Y, Li C, Cui D. Engineered mesenchymal stem cell-derived exosomes with high CXCR4 levels for targeted siRNA gene therapy against cancer. NANOSCALE 2022; 14:4098-4113. [PMID: 35133380 DOI: 10.1039/d1nr08170e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Gene therapy has been used in a variety of diseases and shows brilliant anticancer or cancer suppression effects. Gene therapy is gradually evolving as the most compelling frontier hotspot in the field of cancer therapy. The current vehicles used in gene therapy have poor safety and low delivery efficiency, and thus, it is urgent to develop novel delivery vehicles for gene therapy. Due to the excellent stability and biosafety of exosomes, their use as drug carriers for novel nucleic acid therapy is in full swing, revealing huge prospects for clinical application. Mesenchymal stem cells (MSCs) have a natural homing property and can spontaneously accumulate at injury sites, inflammation sites, and even tumour sites. This feature is attributed to a variety of tropism factors expressed on their surface; for example, CXC chemokine receptor type 4 (CXCR4) can specifically bind to the highly expressed stromal cell derived factor-1 (SDF-1) on the tumour surface, which is essential for accumulation of MSCs at the tumour site. The mesenchymal stem cells used in this study were genetically engineered to obtain exosomes with high CXCR4 expression as carriers for targeted gene-drug delivery, and then, the Survivin gene was loaded via electrotransformation to construct a brand-new gene-drug delivery system (CXCR4high Exo/si-Survivin). Finally, related in vivo and in vitro experiments were conducted. We observed that the new delivery system can efficiently aggregate at the tumour site and release siRNA into tumour cells, knocking down the Survivin gene in tumour cells in vivo and thereby inhibiting tumour growth. This new gene-drug delivery system has tremendous clinical transformation value and provides a new strategy for clinical treatment of tumours.
Collapse
Affiliation(s)
- Shuyue Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| | - Bin Liu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, PR China.
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Junyi Fan
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cuili Xue
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, PR China.
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Yi Lu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, PR China.
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Can Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, PR China.
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| |
Collapse
|
20
|
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: 1.0] [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
|
21
|
Xu WJ, Cai JX, Li YJ, Wu JY, Xiang D. Recent progress of macrophage vesicle-based drug delivery systems. Drug Deliv Transl Res 2022; 12:2287-2302. [PMID: 34984664 DOI: 10.1007/s13346-021-01110-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 12/13/2022]
Abstract
Nanoparticle drug delivery systems (NDDSs) are promising platforms for efficient delivery of drugs. In the past decades, many nanomedicines have received clinical approval and completed translation. With the rapid advance of nanobiotechnology, natural vectors are emerging as novel strategies to carry and delivery nanoparticles and drugs for biomedical applications. Among diverse types of cells, macrophage is of great interest for their essential roles in inflammatory and immune responses. Macrophage-derived vesicles (MVs), including exosomes, microvesicles, and those from reconstructed membranes, may inherit the chemotactic migration ability and high biocompatibility. The unique properties of MVs make them competing candidates as novel drug delivery systems for precision nanomedicine. In this review, the advantages and disadvantages of existing NDDSs and MV-based drug delivery systems (MVDDSs) were compared. Then, we summarized the potential applications of MVDDSs and discuss future perspectives. The development of MVDDS may provide avenues for the treatment of diseases involving an inflammatory process.
Collapse
Affiliation(s)
- Wen-Jie Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Jia-Xin Cai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Yong-Jiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Jun-Yong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China. .,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China.
| |
Collapse
|
22
|
Haiyan C, Mengyuan Z, Yuteng Z, Ziyan L, Pan W, Han L. Recent advances on biomedical applications of bacterial outer membrane vesicles. J Mater Chem B 2022; 10:7384-7396. [DOI: 10.1039/d2tb00683a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoscale and non-self-replicating outer membrane vesicles (OMVs) are naturally secreted by some bacteria with their structures and compositions similar to that of the outer membrane of parental bacteria. With some...
Collapse
|
23
|
Hua C, Zhang Y, Liu Y. Enhanced Anticancer Efficacy of Chemotherapy by Amphiphilic Y-Shaped Polypeptide Micelles. Front Bioeng Biotechnol 2021; 9:817143. [PMID: 35036402 PMCID: PMC8758568 DOI: 10.3389/fbioe.2021.817143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022] Open
Abstract
Although the treatment modalities of cancers are developing rapidly, chemotherapy is still the primary treatment strategy for most solid cancers. The progress in nanotechnology provides an opportunity to upregulate the tumor suppression efficacy and decreases the systemic toxicities. As a promising nanoplatform, the polymer micelles are fascinating nanocarriers for the encapsulation and delivery of chemotherapeutic agents. The chemical and physical properties of amphiphilic co-polymers could significantly regulate the performances of the micellar self-assembly and affect the behaviors of controlled release of drugs. Herein, two amphiphilic Y-shaped polypeptides are prepared by the ring-opening polymerization of cyclic monomer l-leucine N-carboxyanhydride (l-Leu NCA) initiated by a dual-amino-ended macroinitiator poly(ethylene glycol) [mPEG-(NH2)2]. The block co-polypeptides with PLeu8 and PLeu16 segments could form spontaneously into micelles in an aqueous solution with hydrodynamic radii of 80.0 ± 6.0 and 69.1 ± 4.8 nm, respectively. The developed doxorubicin (DOX)-loaded micelles could release the payload in a sustained pattern and inhibit the growth of xenografted human HepG2 hepatocellular carcinoma with decreased systemic toxicity. The results demonstrated the great potential of polypeptide micellar formulations in cancer therapy clinically.
Collapse
Affiliation(s)
- Cong Hua
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | | | | |
Collapse
|
24
|
Wang D, Zhang X, Xu B. PEGylated Doxorubicin Prodrug-Forming Reduction-Sensitive Micelles With High Drug Loading and Improved Anticancer Therapy. Front Bioeng Biotechnol 2021; 9:781982. [PMID: 34869293 PMCID: PMC8640247 DOI: 10.3389/fbioe.2021.781982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/25/2021] [Indexed: 01/29/2023] Open
Abstract
Significant efforts on the design and development of advanced drug delivery systems for targeted cancer chemotherapy continue to be a major challenge. Here, we reported a kind of reduction-responsive PEGylated doxorubicin (DOX) prodrug via the simple esterification and amidation reactions, which self-assembled into the biodegradable micelles in solutions. Since there was an obvious difference in the reduction potentials between the oxidizing extracellular milieu and the reducing intracellular fluids, these PEG-disulfide-DOX micelles were localized intracellularly and degraded rapidly by the stimulus to release the drugs once reaching the targeted tumors, which obviously enhanced the therapeutic efficacy with low side effects. Moreover, these reduction-sensitive micelles could also physically encapsulate the free DOX drug into the polymeric cargo, exhibiting a two-phase programmed drug release behavior. Consequently, it showed a potential to develop an intelligent and multifunctional chemotherapeutic payload transporter for the effective tumor therapy.
Collapse
Affiliation(s)
- Dongdong Wang
- Minimally Invasive Interventional Therapy Center, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
- Department of Oncology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiaoyi Zhang
- School of Pharmacy, Shihezi University, Shihezi, China
| | - Bingbing Xu
- Sports Medicine Department, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
- Institute of Sports Medicine of Peking University, Beijing, China
| |
Collapse
|
25
|
Tang C, Liu H, Fan Y, He J, Li F, Wang J, Hou Y. Functional Nanomedicines for Targeted Therapy of Bladder Cancer. Front Pharmacol 2021; 12:778973. [PMID: 34867408 PMCID: PMC8635105 DOI: 10.3389/fphar.2021.778973] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/29/2021] [Indexed: 12/29/2022] Open
Abstract
Bladder cancer is one of most common malignant urinary tract tumor types with high incidence worldwide. In general, transurethral resection of non-muscle-invasive bladder cancer followed by intravesical instillation of chemotherapy is the standard treatment approach to minimize recurrence and delay progression of bladder cancer. However, conventional intravesical chemotherapy lacks selectivity for tumor tissues and the concentration of drug is reduced with the excretion of urine, leading to frequent administration and heavy local irritation symptoms. While nanomedicines can overcome all the above shortcomings and adhere to the surface of bladder tumors for a long time, and continuously and efficiently release drugs to bladder cancers. The rapid advances in targeted therapy have led to significant improvements in drug efficacy and precision of targeted drug delivery to eradicate tumor cells, with reduced side-effects. This review summarizes the different available nano-systems of targeted drug delivery to bladder cancer tissues. The challenges and prospects of targeted therapy for bladder cancer are additionally discussed.
Collapse
Affiliation(s)
- Chao Tang
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| | - Heng Liu
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| | - Yanpeng Fan
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| | - Jiahao He
- School of Chemical Engineering, Changchun University of Technology, Changchun, China
| | - Fuqiu Li
- Department of Dermatology, the Second Hospital of Jilin University, Changchun, China
| | - Jin Wang
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| | - Yuchuan Hou
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Jiang Z, Li T, Cheng H, Zhang F, Yang X, Wang S, Zhou J, Ding Y. Nanomedicine potentiates mild photothermal therapy for tumor ablation. Asian J Pharm Sci 2021; 16:738-761. [PMID: 35027951 PMCID: PMC8739255 DOI: 10.1016/j.ajps.2021.10.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 12/17/2022] Open
Abstract
The booming photothermal therapy (PTT) has achieved great progress in non-invasive oncotherapy, and paves a novel way for clinical oncotherapy. Of note, mild temperature PTT (mPTT) of 42–45 °C could avoid treatment bottleneck of the traditional PTT, including nonspecific injury to normal tissues, vasculature and host antitumor immunity. However, cancer cells can resist mPTT via heat shock response and autophagy, thus leading to insufficient mPTT monotherapy to ablate tumor. To overcome the deficient antitumor efficacy caused by thermo-resistance of cancer cells and mono mPTT, synergistic therapies towards cancer cells have been conducted with mPTT. This review summarizes the recent advances in nanomedicine-potentiated mPTT for cancer treatment, including strategies for enhanced single-mode mPTT and mPTT plus synergistic therapies. Moreover, challenges and prospects for clinical translation of nanomedicine-potentiated mPTT are discussed.
Collapse
|
27
|
Xu Y, Zhang X, Hu G, Wu X, Nie Y, Wu H, Kong D, Ning X. Multistage targeted "Photoactive neutrophil" for enhancing synergistic photo-chemotherapy. Biomaterials 2021; 279:121224. [PMID: 34710792 DOI: 10.1016/j.biomaterials.2021.121224] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/26/2021] [Accepted: 10/22/2021] [Indexed: 01/04/2023]
Abstract
Cell-based drug delivery system holds a great promise in anticancer treatment, due to its potential of maximizing therapeutic efficacy while minimizing adverse effects. However, current cell system can only deliver drugs in tumor lesions, but lack an ability to target subcellular locus of therapeutic actions, thereby compromising anticancer efficacy. Herein, we bioengineered living neutrophils as a novel type of "Photoactive neutrophil" (PAN) with capabilities of self-amplified multistage targeting and inflammation response for enhancing mitochondria-specific photo-chemotherapy. PAN encapsulated multifunctional nanocomplex (RA/Ce6) of RGD-apoptotic peptide conjugate (RA) decorated liposomal photosensitizer Ce6, and could overcome tumor barriers to selectively release RA/Ce6 within tumor. Consequently, RA/Ce6 actively entered cancer cells and accumulated in mitochondria to trigger combined photodynamic therapy (PDT) and RA-induced mitochondrial membrane disruption, resulting in enhanced therapeutic effects. Importantly, PAN exhibited inflammation amplified tumor targeting after PDT, and initiated combined photo-chemotherapy to suppress tumor growth without adverse effects, leading to prolonged mice survival. Therefore, PAN represents the first multistage targeted cell therapy, and brings new insights into cancer treatment.
Collapse
Affiliation(s)
- Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Xiaomin Zhang
- Department of Pediatric Stomatology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Getian Hu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| | - Xiaotong Wu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Yuanyuan Nie
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Heming Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China.
| | - Desheng Kong
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China.
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
28
|
|
29
|
Chauhan A, Khan T, Omri A. Design and Encapsulation of Immunomodulators onto Gold Nanoparticles in Cancer Immunotherapy. Int J Mol Sci 2021; 22:8037. [PMID: 34360803 PMCID: PMC8347387 DOI: 10.3390/ijms22158037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
The aim of cancer immunotherapy is to reactivate autoimmune responses to combat cancer cells. To stimulate the immune system, immunomodulators, such as adjuvants, cytokines, vaccines, and checkpoint inhibitors, are extensively designed and studied. Immunomodulators have several drawbacks, such as drug instability, limited half-life, rapid drug clearance, and uncontrolled immune responses when used directly in cancer immunotherapy. Several strategies have been used to overcome these limitations. A simple and effective approach is the loading of immunomodulators onto gold-based nanoparticles (GNPs). As gold is highly biocompatible, GNPs can be administered intravenously, which aids in increasing cancer cell permeability and retention time. Various gold nanoplatforms, including nanospheres, nanoshells, nanorods, nanocages, and nanostars have been effectively used in cancer immunotherapy. Gold nanostars (GNS) are one of the most promising GNP platforms because of their unusual star-shaped geometry, which significantly increases light absorption and provides high photon-to-heat conversion efficiency due to the plasmonic effect. As a result, GNPs are a useful vehicle for delivering antigens and adjuvants that support the immune system in killing tumor cells by facilitating or activating cytotoxic T lymphocytes. This review represents recent progress in encapsulating immunomodulators into GNPs for utility in a cancer immunotherapeutic regimen.
Collapse
Affiliation(s)
- Akshita Chauhan
- Department of Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, Maharashtra, India;
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, Maharashtra, India;
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
30
|
Yang T, Wang D, Chen X, Liang Y, Guo F, Wu C, Jia L, Hou Z, Li W, He Z, Wang X. 18F-ASEM Imaging for Evaluating Atherosclerotic Plaques Linked to α7-Nicotinic Acetylcholine Receptor. Front Bioeng Biotechnol 2021; 9:684221. [PMID: 34277585 PMCID: PMC8280778 DOI: 10.3389/fbioe.2021.684221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022] Open
Abstract
Background Atherosclerosis is a chronic vascular inflammatory procedure alongside with lipid efflux disorder and foam cell formation. α7-Nicotinic acetylcholine receptor (α7nAChR) is a gated-calcium transmembrane channel widely expressed in neuron and non-neuron cells, such as monocytes and macrophages, activated T cells, dendritic cells, and mast cells. 18F-ASEM is an inhibitor targeted to α7nAChR that had been successfully applied in nervous system diseases. Previous studies had highlighted that α7nAChR was related to the emergency of vulnerable atherosclerotic plaques with excess inflammation cells. Thus, 18F-ASEM could be a complementary diagnostic approach to atherosclerotic plaques. Materials and Methods The synthesis of ASEM precursor and 18F-labeling had been performed successfully. We had established the ApoE–/– mice atherosclerotic plaques model (fed with western diet) and New Zealand rabbits atherosclerotic models (balloon-sprained experiment and western diet). After damage of endothelial cells and primary plaque formation, 18F-ASEM imaging of atherosclerotic plaques linked to α7nAChR had been conducted. In vivo micro-PET/CT imaging of ApoE–/– mice and the control group was performed 1 h after injection of 18F-ASEM (100–150 μCi); PET/CT imaging for rabbits with atherosclerotic plaques and control ones was also performed. Meanwhile, we also conducted CT scan on the abdominal aorta of these rabbits. After that, the animals were sacrificed, and the carotid and abdominal aorta were separately taken out for circular sections. The paraffin-embedded specimens were sectioned with 5 μm thickness and stained with hematoxylin–eosin (H&E) and oil red. Results In vivo vessel binding of 18F-ASEM and α7nAChR expression in the model group with atherosclerosis plaques was significantly higher than that in the control group. PET/CT imaging successfully identified the atherosclerotic plaques in ApoE–/– mice and model rabbits, whereas no obvious signals were detected in normal mice or rabbits. Compared with 18F-FDG, 18F-ASEM had more significant effect on the early monitoring of inflammation in carotid atherosclerotic plaques of ApoE–/– mice and model rabbits. 18F-ASEM had relatively more palpable effect on the imaging of abdominal aorta with atherosclerosis in rabbits. H&E and oil red staining identified the formation of atherosclerotic plaques in model animals, which provided pathological basis for the evaluation of imaging effects. Conclusion We first confirmed 18F-ASEM as radiotracer with good imaging properties for precise identification of atherosclerotic diseases.
Collapse
Affiliation(s)
- Tao Yang
- Department of Cardiovascular Surgery, Fu Wai Hospital, Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dawei Wang
- Department of Nuclear Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Xiangyi Chen
- Department of Nuclear Medicine, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China.,Yichang Central People's Hospital, Yichang, China
| | - Yingkui Liang
- Department of Nuclear Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Feng Guo
- Department of Nuclear Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Chunxiao Wu
- Department of Cardiovascular Surgery, Fu Wai Hospital, Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liujun Jia
- Department of Cardiovascular Surgery, Fu Wai Hospital, Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihui Hou
- Department of Cardiovascular Surgery, Fu Wai Hospital, Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenliang Li
- School of Pharmacy, Jilin Medical University, Jilin City, China
| | - ZuoXiang He
- Department of Nuclear Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Xin Wang
- Department of Cardiovascular Surgery, Fu Wai Hospital, Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Wang F, Ullah A, Fan X, Xu Z, Zong R, Wang X, Chen G. Delivery of nanoparticle antigens to antigen-presenting cells: from extracellular specific targeting to intracellular responsive presentation. J Control Release 2021; 333:107-128. [PMID: 33774119 DOI: 10.1016/j.jconrel.2021.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/05/2023]
Abstract
An appropriate delivery system can improve the immune effects of antigens against various infections or tumors. Antigen-presenting cells (APCs) are specialized to capture and process antigens in vivo, which link the innate and adaptive immune responses. Functionalization of vaccine delivery systems with targeting moieties to APCs is a promising strategy for provoking potent immune responses. Additionally, the internalization and intracellular distribution of antigens are closely related to the initiation of downstream immune responses. With a deeper understanding of the intracellular microenvironment and the mechanisms of antigen presentation, vehicles designed to respond to endogenous and external stimuli can modulate antigen processing and presentation pathways, which are critical to the types of immune response. Here, an overview of extracellular targeting delivery of antigens to APCs and intracellular stimulus-responsiveness strategies is provided, which might be helpful for the rational design of vaccine delivery systems.
Collapse
Affiliation(s)
- Fei Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Aftab Ullah
- Shantou University Medical College, Shantou 515041, China
| | - Xuelian Fan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Zhou Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Rongling Zong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xuewen Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Gang Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
32
|
Zhang H, Zhang J, Liu Y, Jiang Y, Li Z. Molecular Targeted Agent and Immune Checkpoint Inhibitor Co-Loaded Thermosensitive Hydrogel for Synergistic Therapy of Rectal Cancer. Front Pharmacol 2021; 12:671611. [PMID: 33935796 PMCID: PMC8085774 DOI: 10.3389/fphar.2021.671611] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/26/2021] [Indexed: 01/04/2023] Open
Abstract
Molecular targeted therapy has been proved effective in treatment of rectal cancer. Up-regulated expression of programmed death ligand-1 (PD-L1) was observed after the management of molecular targeted therapy, which made the therapeutic effect discounted. Tumors with higher PD-L1 expression were more sensitive and responsive to treatment of PD-L1 inhibitor. Therefore, the combination of molecular targeted therapy and immune checkpoint blockade makes sense. In this study, the copolymers of poly (ethylene glycol)-block-poly (L-leucine) (PEG-PLLeu) were synthesized as a thermosensitive hydrogel composite for consecutive release of regorafenib (REG) and BMS202. The mechanical properties of PEG-PLLeu were investigated, confirming that PEG-PLLeu (5 wt.%) was suitable for in situ injection as drug-delivery composite at low temperature and stable after sol-gel transition at body temperature. Importantly, the double drug loaded hydrogel showed superior antitumour activity over single drugs in an orthotopic rectal cancer model (CT26-Luc). Further analysis of the tumor tissues suggested that REG upregulated the expression of PD-L1 in tumor tissues. In addition, the immunosuppressive tumor microenvironment of CT26-Luc tumor was distinctly relieved under the effect of BMS202, as characterized by increased infiltration of CD8+ T cells in tumors and enhanced secretion of antitumour cytokines (IFN-γ and TNF-α). Moreover, the drug-loaded composite showed no obvious toxicity in histological analysis. Taken together, the administration of REG and BMS202 in the PEG-PLLeu composite could induce a synergistic effect in in situ treatment of rectal cancer without obvious toxicity, and thus represented a potential strategy for enhanced in situ therapeutic modality.
Collapse
Affiliation(s)
- Huaiyu Zhang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiayu Zhang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yilun Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Jiang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
33
|
Liu J, Li Z, Zhao D, Feng X, Wang C, Li D, Ding J. Immunogenic cell death-inducing chemotherapeutic nanoformulations potentiate combination chemoimmunotherapy. MATERIALS & DESIGN 2021; 202:109465. [DOI: 10.1016/j.matdes.2021.109465] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
|
34
|
Choi JS, Park JW, Seu YB, Doh KO. Enhanced efficacy of folate-incorporated cholesteryl doxorubicin liposome in folate receptor abundant cancer cell. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
35
|
Gao J, Huang J, Shi R, Wei J, Lei X, Dou Y, Li Y, Zuo Y, Li J. Loading and Releasing Behavior of Selenium and Doxorubicin Hydrochloride in Hydroxyapatite with Different Morphologies. ACS OMEGA 2021; 6:8365-8375. [PMID: 33817497 PMCID: PMC8015115 DOI: 10.1021/acsomega.1c00092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/05/2021] [Indexed: 02/08/2023]
Abstract
![]()
Doxorubicin (Dox)-loaded
or selenium-substituted hydroxyapatite
(HA) has been developed to achieve anti-osteosarcoma or bone regeneration
in a number of studies. However, currently, there is a lack of studies
on the combination of Dox and selenium loading in/on HA and comparative
research studies on which form and size of HA are more suitable for
drug loading and release in the treatment osteogenesis after osteosarcoma
resection. Herein, selenium-doped rod-shaped nano-HA (n-HA) and spherical
mesoporous HA (m-HA) were successfully prepared. The doping efficiency
of selenium and the Dox loading capacity of selenium-doped HA with
different morphologies were studied. The release kinetics of Dox and
the selenium element in phosphate-buffered saline with different pH
values was also comparatively investigated. The drug loading results
showed that n-HA exhibited 3 times higher selenium doping amount than
m-HA, and the Dox entrapment efficiency of selenium-doped n-HA (0.1Se-n-HA)
presented 20% higher than that of selenium-doped m-HA (0.1Se-m-HA).
The Dox release behaviors of HA in two different morphologies showed
similar release kinetics, with almost the same Dox releasing ratio
but slightly more Dox releasing amount in selenium-doped HA than in
HA without selenium. The selenium release from selenium-doped n-HA-D
(0.1Se-n-HA-D) particles was 2 times as much as that of selenium-doped
m-HA-D (0.1Se-m-HA) particles. Our study indicated that n-HA loaded
with Dox and selenium may be a promising drug delivery strategy for
inhibition of osteosarcoma recurrence and promoting osteogenesis simultaneously.
Collapse
Affiliation(s)
- Jing Gao
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Jinhui Huang
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Rui Shi
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jiawei Wei
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Xiaoyu Lei
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Yichen Dou
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Yubao Li
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Yi Zuo
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Jidong Li
- Research Center for Nano Biomaterials, Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| |
Collapse
|
36
|
Song J, Xu B, Yao H, Lu X, Tan Y, Wang B, Wang X, Yang Z. Schiff-Linked PEGylated Doxorubicin Prodrug Forming pH-Responsive Nanoparticles With High Drug Loading and Effective Anticancer Therapy. Front Oncol 2021; 11:656717. [PMID: 33842372 PMCID: PMC8027505 DOI: 10.3389/fonc.2021.656717] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/05/2021] [Indexed: 12/28/2022] Open
Abstract
Developing efficacious drug delivery systems for targeted cancer chemotherapy remains a major challenge. Here we demonstrated a kind of pH-responsive PEGylated doxorubicin (DOX) prodrug via the effective esterification and Schiff base reactions, which could self-assemble into the biodegradable micelles in aqueous solutions. Owing to low pH values inside the tumor cells, these PEG-Schiff-DOX nanoparticles exhibited high drug loading ability and pH-responsive drug release behavior within the tumor cells or tissues upon changes in physical and chemical environments, but they displayed good stability at physiological conditions for a long period. CCK-8 assay showed that these PEGylated DOX prodrugs had a similar cytotoxicity to the MCF-7 tumor cells as the free DOX drug. Moreover, this kind of nanoparticle could also encapsulate small DOX drugs with high drug loading, sufficient drug release and enhanced therapeutic effects toward MCF-7 cells, which will be benefited for developing more drug carriers with desirable functions for clinical anticancer therapy.
Collapse
Affiliation(s)
- Jian Song
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Bingbing Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
| | - Hui Yao
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaofang Lu
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Yang Tan
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Bingyang Wang
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zheng Yang
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
37
|
Li L, Tong T, Ji Q, Xu Z, Guan Y, Liang X, Huang H, Kang Y, Pang J. Dual pH- and Glutathione-Responsive CO 2-Generating Nanodrug Delivery System for Contrast-Enhanced Ultrasonography and Therapy of Prostate Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:12899-12911. [PMID: 33720701 DOI: 10.1021/acsami.1c00077] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Ultrasonography (US) contrast imaging using US contrast agents has been widely applied for the diagnosis and differential diagnosis of tumors. Commercial US contrast agents have limited applications because of their large size and shorter imaging time. At the same time, the desired therapeutic purpose cannot be achieved by applying only conventional US contrast agents. The development of nanoscale US agents with US imaging and therapeutic functions has attracted increasing attention. In this study, we successfully developed DOX-loaded poly-1,6-hexanedithiol-sodium bicarbonate nanoparticles (DOX@HADT-SS-NaHCO3 NPs) with pH-responsive NaHCO3 and GSH-responsive disulfide linkages. DOX@HADT-SS-NaHCO3 NPs underwent acid-triggered decomposition of NaHCO3 to generate CO2 bubbles and a reduction of disulfide linkages to further promote the release of CO2 and DOX. The potential of DOX@HADT-SS-NaHCO3 NPs for contrast-enhanced US imaging and therapy of prostate cancer was thoroughly evaluated using in vitro agarose gel phantoms and a C4-2 tumor-bearing nude mice model. These polymeric NPs displayed significantly enhanced US contrast at acidic pH and antitumor efficacy. Therefore, the NaHCO3 and DOX-encapsulated polymeric NPs hold tremendous potential for effective US imaging and therapy of prostate cancer.
Collapse
Affiliation(s)
- Lujing Li
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
- Department of Ultrasound, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Tongyu Tong
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Qiao Ji
- Department of Ultrasound, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Zuofeng Xu
- Department of Ultrasound, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yupeng Guan
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xin Liang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Hai Huang
- Department of Urology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yang Kang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
38
|
Wang D, Yao Y, Wang S, Zhang H, He ZX. The Availability of the α7-Nicotinic Acetylcholine Receptor in Early Identification of Vulnerable Atherosclerotic Plaques: A Study Using a Novel 18F-Label Radioligand PET. Front Bioeng Biotechnol 2021; 9:640037. [PMID: 33777911 PMCID: PMC7994753 DOI: 10.3389/fbioe.2021.640037] [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: 12/10/2020] [Accepted: 01/14/2021] [Indexed: 11/17/2022] Open
Abstract
Background: It has been confirmed that the α7-nicotinic acetylcholine receptor (α7nAChR) is an important target for identifying vulnerable atherosclerotic plaques. Previously, we successfully designed and synthesized a series of 18F-labeled PET molecular probes targeting α7nAChR, which are mainly used in the diagnosis of Alzheimer's disease. Based on the characteristics of α7nAChR in blood vessels, we have firstly screened for a suitable novel 18F-labeled PET molecular probe ([18F]YLF-DW), with high selectivity for α7nAChR over α4β2nAChR and a good effect for the imaging of atherosclerotic animal models, to effectively identify vulnerable atherosclerotic plaques at an early stage. Meanwhile, we compared it with the “gold standard” pathological examination of atherosclerosis, to verify the reliability of [18F]YLF-DW in early diagnosis of atherosclerosis. Methods: The vulnerable atherosclerotic plaques model of ApoE-/-mice were successfully established. Then based on the methods of 3D-QSAR and molecular docking, we designed oxazolo[4,5-b] pyridines and fluorenone compounds, which are targeted at α7nAChR. Through further screening, a novel alpha7 nicotinic acetylcholine receptor radioligand ([18F]YLF-DW) was synthesized and automatically 18F-labeled using a Stynthra RNplus module. Subsequently, we employed [18F]YLF-DW for the targeting of α7nAChR in atherosclerotic plaques and control group, using a micro-PET/CT respectively. After imaging, the mice were sacrificed by air embolism and the carotid arteries taken out for making circular sections. The paraffin embedded specimens were sectioned with 5 μm thickness and stained with oil red. After staining, immunohistochemistry experiment was carried out to verify the effect of micro-PET/CT imaging. Results: The micro-PET/CT imaging successfully identified the vulnerable atherosclerotic plaques in the carotid arteries of ApoE-/-mice; whereas, no signal was observed in normal control mice. In addition, compared with the traditional imaging agent [18F]FDG, [18F]YLF-DW had a significant effect on the early plaques imaging of carotid atherosclerosis. The results of oil red staining and immunohistochemistry also showed early formations of carotid plaques in ApoE-/-mice and provided pathological bases for the evaluation of imaging effect. Conclusion: We innovated to apply the novel molecular probe ([18F]YLF-DW) to the identification of vulnerable atherosclerotic plaques in carotid arteries, to detect atherosclerosis early inflammatory response and provide powerful input for the early diagnosis of atherosclerotic lesions, which may play an early warning role in cardiovascular acute events.
Collapse
Affiliation(s)
- Dawei Wang
- State Key Laboratory of Cardiovascular Disease, Department of Nuclear Medicine, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Yao
- State Key Laboratory of Cardiovascular Disease, Department of Nuclear Medicine, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuxia Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Huabei Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Zuo-Xiang He
- Department of Nuclear Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| |
Collapse
|
39
|
Lu L, Sun Y, Wan C, Hu Y, Lo PC, Lovell JF, Yang K, Jin H. Role of intravital imaging in nanomedicine-assisted anti-cancer therapy. Curr Opin Biotechnol 2021; 69:153-161. [PMID: 33476937 DOI: 10.1016/j.copbio.2020.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 01/07/2023]
Abstract
Although nanomedicines have provided promising anti-tumor effects in cancer animal models, their clinical success remains limited. One of the most significant barriers in the clinical translation of nanomedicines is that they consist of multiple components, each of which may have different toxicities and therapeutic effects. Intravital imaging provides high spatial and temporal resolution for visualizing nanomedicine-mediated interactions between immune cells and tumor cells in real-time. Intravital imaging can facilitate the in vivo evaluation of the properties and effects of nanomedicines, such as their ability to cross the tumor vasculature, specifically eliminate the cancer cells, and modulate the immune cells found in the tumor microenvironment (TME). Thus, intravital imaging can provide direct evidence of nanomedicine's intravital behavior to better understand mechanism and accelerate clinical translation. In this review, we summarize several applications and latest advances in intravital imaging in nanomedicine-assisted anti-cancer therapy and discuss future perspectives in the field.
Collapse
Affiliation(s)
- Lisen Lu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
40
|
Alshehri S, Imam SS, Rizwanullah M, Akhter S, Mahdi W, Kazi M, Ahmad J. Progress of Cancer Nanotechnology as Diagnostics, Therapeutics, and Theranostics Nanomedicine: Preclinical Promise and Translational Challenges. Pharmaceutics 2020; 13:E24. [PMID: 33374391 PMCID: PMC7823416 DOI: 10.3390/pharmaceutics13010024] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Early detection, right therapeutic intervention, and simultaneous effectiveness mapping are considered the critical factors in successful cancer therapy. Nevertheless, these factors experience the limitations of conventional cancer diagnostics and therapeutics delivery approaches. Along with providing the targeted therapeutics delivery, advances in nanomedicines have allowed the combination of therapy and diagnostics in a single system (called cancer theranostics). This paper discusses the progress in the pre-clinical and clinical development of therapeutics, diagnostics, and theranostics cancer nanomedicines. It has been well evident that compared to the overabundance of works that claimed success in pre-clinical studies, merely 15 and around 75 cancer nanomedicines are approved, and currently under clinical trials, respectively. Thus, we also brief the critical bottlenecks in the successful clinical translation of cancer nanomedicines.
Collapse
Affiliation(s)
- Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.); (W.M.); (M.K.)
- Department of Pharmaceutical Sciences, College of Pharmacy, Almaarefa University, Riyadh 11597, Saudi Arabia
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.); (W.M.); (M.K.)
| | - Md. Rizwanullah
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India; or
| | - Sohail Akhter
- New Product Development, Global R&D, Sterile ops, TEVA Pharmaceutical Industries Ltd., Aston Ln N, Halton, Preston Brook, Runcorn WA7 3FA, UK;
| | - Wael Mahdi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.); (W.M.); (M.K.)
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.); (W.M.); (M.K.)
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| |
Collapse
|
41
|
Zhao Z, Li D, Wu Z, Wang Q, Ma Z, Zhang C. Research Progress and Prospect of Nanoplatforms for Treatment of Oral Cancer. Front Pharmacol 2020; 11:616101. [PMID: 33391000 PMCID: PMC7773899 DOI: 10.3389/fphar.2020.616101] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/30/2020] [Indexed: 12/27/2022] Open
Abstract
Oral cancers refer to malignant tumors associated with high morbidity and mortality, and oral squamous cell carcinoma accounts for the majority of cases. It is an important part of head and neck, and oral cancer is one of the six most common cancers in the world. At present, the traditional treatment methods for oral cancer include surgery, radiation therapy, and chemotherapy. However, these methods have many disadvantages. In recent years, nanomedicine, the delivery of drugs through nanoplatforms for the treatment of cancer, has become a promising substitutive therapy. The use of nanoplatforms can reduce the degradation of the drug in the body and accurately deliver it to the tumor site. This minimizes the distribution of the drug to other organs, thereby reducing its toxicity and allowing higher drug concentration at the tumor site. This review introduces polymer nanoparticles, lipid-based nanoparticles, metal nanoparticles, hydrogels, exosomes, and dendrimers for the treatment of oral cancer, and discusses how these nanoplatforms play an anti-cancer effect. Finally, the review gives a slight outlook on the future prospects of nanoplatforms for oral cancer treatment.
Collapse
Affiliation(s)
- Zhilong Zhao
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| | - Dan Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Ziqi Wu
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| | - Qihui Wang
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| | | | - Congxiao Zhang
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
42
|
Li J, Luo Y, Li B, Xia Y, Wang H, Fu C. Implantable and Injectable Biomaterial Scaffolds for Cancer Immunotherapy. Front Bioeng Biotechnol 2020; 8:612950. [PMID: 33330440 PMCID: PMC7734317 DOI: 10.3389/fbioe.2020.612950] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer immunotherapy has become an emerging strategy recently producing durable immune responses in patients with varieties of malignant tumors. However, the main limitation for the broad application of immunotherapies still to reduce side effects by controlling and regulating the immune system. In order to improve both efficacy and safety, biomaterials have been applied to immunotherapies for the specific modulation of immune cells and the immunosuppressive tumor microenvironment. Recently, researchers have constantly developed biomaterials with new structures, properties and functions. This review provides the most recent advances in the delivery strategies of immunotherapies based on localized biomaterials, focusing on the implantable and injectable biomaterial scaffolds. Finally, the challenges and prospects of applying implantable and injectable biomaterial scaffolds in the development of future cancer immunotherapies are discussed.
Collapse
Affiliation(s)
- Jie Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yiqian Luo
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Baoqin Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Isolation and characterization of nanocrystalline cellulose from flaxseed Hull: A future onco-drug delivery agent. JOURNAL OF SAUDI CHEMICAL SOCIETY 2020. [DOI: 10.1016/j.jscs.2020.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
44
|
Hamedani Y, Chakraborty S, Sabarwal A, Pal S, Bhowmick S, Balan M. Novel Honokiol-eluting PLGA-based scaffold effectively restricts the growth of renal cancer cells. PLoS One 2020; 15:e0243837. [PMID: 33332399 PMCID: PMC7746163 DOI: 10.1371/journal.pone.0243837] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Renal Cell Carcinoma (RCC) often becomes resistant to targeted therapies, and in addition, dose-dependent toxicities limit the effectiveness of therapeutic agents. Therefore, identifying novel drug delivery approaches to achieve optimal dosing of therapeutic agents can be beneficial in managing toxicities and to attain optimal therapeutic effects. Previously, we have demonstrated that Honokiol, a natural compound with potent anti-tumorigenic and anti-inflammatory effects, can induce cancer cell apoptosis and inhibit the growth of renal tumors in vivo. In cancer treatment, implant-based drug delivery systems can be used for gradual and sustained delivery of therapeutic agents like Honokiol to minimize systemic toxicity. Electrospun polymeric fibrous scaffolds are ideal candidates to be used as drug implants due to their favorable morphological properties such as high surface to volume ratio, flexibility and ease of fabrication. In this study, we fabricated Honokiol-loaded Poly(lactide-co-glycolide) (PLGA) electrospun scaffolds; and evaluated their structural characterization and biological activity. Proton nuclear magnetic resonance data proved the existence of Honokiol in the drug loaded polymeric scaffolds. The release kinetics showed that only 24% of the loaded Honokiol were released in 24hr, suggesting that sustained delivery of Honokiol is feasible. We calculated the cumulative concentration of the Honokiol released from the scaffold in 24hr; and the extent of renal cancer cell apoptosis induced with the released Honokiol is similar to an equivalent concentration of direct application of Honokiol. Also, Honokiol-loaded scaffolds placed directly in renal cell culture inhibited renal cancer cell proliferation and migration. Together, we demonstrate that Honokiol delivered through electrospun PLGA-based scaffolds is effective in inhibiting the growth of renal cancer cells; and our data necessitates further in vivo studies to explore the potential of sustained release of therapeutic agents-loaded electrospun scaffolds in the treatment of RCC and other cancer types.
Collapse
Affiliation(s)
- Yasaman Hamedani
- Department of Mechanical Engineering, Biomedical Engineering and Biotechnology Program, University of Massachusetts Dartmouth, Dartmouth, MA, United States of America
| | - Samik Chakraborty
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Akash Sabarwal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Soumitro Pal
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Sankha Bhowmick
- Department of Mechanical Engineering, Biomedical Engineering and Biotechnology Program, University of Massachusetts Dartmouth, Dartmouth, MA, United States of America
- * E-mail: (MB); (SB)
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
- * E-mail: (MB); (SB)
| |
Collapse
|