51
|
Nasirmoghadas P, Mousakhani A, Behzad F, Beheshtkhoo N, Hassanzadeh A, Nikoo M, Mehrabi M, Kouhbanani MAJ. Nanoparticles in cancer immunotherapies: An innovative strategy. Biotechnol Prog 2020; 37:e3070. [PMID: 32829506 DOI: 10.1002/btpr.3070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022]
Abstract
Cancer has been one of the most significant causes of mortality, worldwide. Cancer immunotherapy has recently emerged as a competent, cancer-fighting clinical strategy. Nevertheless, due to the difficulty of such treatments, costs, and off-target adverse effects, the implementation of cancer immunotherapy described by the antigen-presenting cell (APC) vaccine and chimeric antigen receptor T cell therapy ex vivo in large clinical trials have been limited. Nowadays, the nanoparticles theranostic system as a promising target-based modality provides new opportunities to improve cancer immunotherapy difficulties and reduce their adverse effects. Meanwhile, the appropriate engineering of nanoparticles taking into consideration nanoparticle characteristics, such as, size, shape, and surface features, as well as the use of these physicochemical properties for suitable biological interactions, provides new possibilities for the application of nanoparticles in cancer immunotherapy. In this review article, we focus on the latest state-of-the-art nanoparticle-based antigen/adjuvant delivery vehicle strategies to professional APCs and engineering specific T lymphocyte required for improving the efficiency of tumor-specific immunotherapy.
Collapse
Affiliation(s)
- Pourya Nasirmoghadas
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akbar Mousakhani
- Department of Plant Sciences, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Farahnaz Behzad
- Research Institute for Fundamental Sciences (RIFS), University of Tabriz, Tabriz, Iran
| | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Helal Iran Pharmaceutical and Clinical Complex, Tehran, Iran
| | - Mohsen Mehrabi
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
52
|
Hypoxia-sensitive micellar nanoparticles for co-delivery of siRNA and chemotherapeutics to overcome multi-drug resistance in tumor cells. Int J Pharm 2020; 590:119915. [DOI: 10.1016/j.ijpharm.2020.119915] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/15/2020] [Accepted: 09/20/2020] [Indexed: 12/20/2022]
|
53
|
Luo YL, Liang LF, Gan YJ, Liu J, Zhang Y, Fan YN, Zhao G, Czarna A, Lu ZD, Du XJ, Shen S, Xu CF, Lian ZX, Wang J. An All-in-One Nanomedicine Consisting of CRISPR-Cas9 and an Autoantigen Peptide for Restoring Specific Immune Tolerance. ACS APPLIED MATERIALS & INTERFACES 2020; 12:48259-48271. [PMID: 33070614 DOI: 10.1021/acsami.0c10885] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanotechnology has shown great promise in treating diverse diseases. However, developing nanomedicines that can cure autoimmune diseases without causing systemic immunosuppression is still quite challenging. Herein, we propose an all-in-one nanomedicine comprising an autoantigen peptide and CRISPR-Cas9 to restore specific immune tolerance by engineering dendritic cells (DCs) into a tolerogenic phenotype, which can expand autoantigen-specific regulatory T (Treg) cells. In brief, we utilized cationic lipid-assisted poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-PLGA) nanoparticles to simultaneously encapsulate an autoimmune diabetes-relevant peptide (2.5mi), a CRISPR-Cas9 plasmid (pCas9), and three guide RNAs (gRNAs) targeting costimulatory molecules (CD80, CD86, and CD40). We demonstrated that the all-in-one nanomedicine was able to effectively codeliver these components into DCs, followed by simultaneous disruption of the three costimulatory molecules and presentation of the 2.5mi peptide on the genome-edited DCs. The resulting tolerogenic DCs triggered the generation and expansion of autoantigen-specific Treg cells by presenting the 2.5mi peptide to CD4+ T cells in the absence of costimulatory signals. Using autoimmune type 1 diabetes (T1D) as a typical disease model, we demonstrated that our nanomedicine prevented autoimmunity to islet components and inhibited T1D development. Our all-in-one nanomedicine achieved codelivery of CRISPR-Cas9 and the peptide to DCs and could be easily applied to other autoimmune diseases by substitution of different autoantigen peptides.
Collapse
Affiliation(s)
- Ying-Li Luo
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Li-Fang Liang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yun-Jiu Gan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, P. R. China
| | - Jing Liu
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yue Zhang
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Ya-Nan Fan
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Gui Zhao
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Anna Czarna
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zi-Dong Lu
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xiao-Jiao Du
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Song Shen
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Cong-Fei Xu
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zhe-Xiong Lian
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jun Wang
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, P. R. China
- Research Institute for Food Nutrition and Human Health, Guangzhou 510641, P. R. China
| |
Collapse
|
54
|
Li L, Li H, Xue J, Chen P, Zhou Q, Zhang C. Nanoparticle-Mediated Simultaneous Downregulation of Placental Nrf2 and sFlt1 Improves Maternal and Fetal Outcomes in a Preeclampsia Mouse Model. ACS Biomater Sci Eng 2020; 6:5866-5873. [PMID: 33320575 DOI: 10.1021/acsbiomaterials.0c00826] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Preeclampsia has impacted 3-5% pregnancies among the world and its complications lead to both maternal and fetal morbidity and mortality. However, management of preeclampsia is limited. Nanoparticles targeting chondroitin sulfate A (CSA) can deliver drugs to placenta. Inactivation of soluble fms-like tyrosine kinase (sFlt-1) and nuclear factor-erythroid 2-like 2 (Nrf2) has been proved to alleviate preeclampsia and improve maternal and fetal outcomes. Carboxyl-polyethylene glycol-poly (d,l-lactide) (COOH-PEG5K-PLA8K), cationic lipid DOTAP, and siNrf2 and sisFlt-1 were used to construct the nanoparticles and conjugating peptides targeting CSA was fabricated to it. The expression levels of proteins and RNAs were estimated by qRT-PCR and Western blot assays. ELISA assays were performed to evaluate levels of circulating sFlt-1. The nanoparticles containing siNrf2 and sisFlt-1 are targeted to the placenta trophoblasts and downregulated the expression levels of Nrf2 and sFlt-1 as well as their downstream genes in the placental cells of model mice. Treatment of nanoparticles induced the expression of angiogenic factors in placenta. Knocking down Nrf2 and sFlt-1 synchronously alleviated the preeclampsia and increased the maternal and fetal outcomes in preeclampsia model mice. Nanoparticle-mediated simultaneous downregulation of placental Nrf2 and sFlt1 improved maternal and fetal outcomes in a preeclampsia mouse model.
Collapse
Affiliation(s)
- Lei Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.,Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China.,Key Laboratory of Birth Regulation and Control Technology of National Health and Family Planning Commission of China, Jinan, Shandong 250025, China
| | - Hongyan Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.,Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Jing Xue
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.,Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Qian Zhou
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.,Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Chunhua Zhang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.,Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| |
Collapse
|
55
|
Li L, Guan Y, Xiong H, Deng T, Ji Q, Xu Z, Kang Y, Pang J. Fundamentals and applications of nanoparticles for ultrasound‐based imaging and therapy. NANO SELECT 2020. [DOI: 10.1002/nano.202000035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Lujing Li
- Department of Urology The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen Guangdong 518107 China
| | - Yupeng Guan
- Department of Urology The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen Guangdong 518107 China
| | - Haiyun Xiong
- Department of Urology The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen Guangdong 518107 China
| | - Tian Deng
- Department of Stomatology 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 Guangdong 518107 China
| | - Zuofeng Xu
- Department of Ultrasound The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen Guangdong 518107 China
| | - Yang Kang
- Department of Urology The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen Guangdong 518107 China
| | - Jun Pang
- Department of Urology The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen Guangdong 518107 China
| |
Collapse
|
56
|
Nanoparticle-facilitated delivery of BAFF-R siRNA for B cell intervention and rheumatoid arthritis therapy. Int Immunopharmacol 2020; 88:106933. [PMID: 32866781 DOI: 10.1016/j.intimp.2020.106933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/06/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
The present study was designed to explore the effects of B-cell activating factor receptor (BAFF-R) siRNA encapsulated nanoparticles on collagen-induced arthritis (CIA). BAFF-R siRNA encapsulated nanoparticles (NP-siBAFF-R) were constructed using a double emulsion method and was characterized by dynamic light scattering and transmission electron microscopy. Cellular uptake of nanoparticles was determined using flow cytometry. The CIA mouse model was established and the mice were intravenously injected with nanoparticles. NP-siBAFF-R effectively decreased the expression of BAFF-R in B cells and facilitated the delivery of siRNA into B cells. Treatment of NPsiBAFF-R ameliorated rheumatoid arthritis (RA) symptoms in the CIA mouse model via decreasing the arthritis score, mean ankle diameter, the levels of anti-collagen IgG in serum and increasing the expression of collagen type II and osteocalcin in dissected joint tissues. Additionally, treatment of NPsiBAFF-R decreased the percentage and number of B cells and inhibited the production of pro-inflammatory cytokines in RA mice. These results demonstrate that NP-siBAFF-R may provide an effective strategy for RA treatment.
Collapse
|
57
|
Wei W, Sun J, Guo XY, Chen X, Wang R, Qiu C, Zhang HT, Pang WH, Wang JC, Zhang Q. Microfluidic-Based Holonomic Constraints of siRNA in the Kernel of Lipid/Polymer Hybrid Nanoassemblies for Improving Stable and Safe In Vivo Delivery. ACS APPLIED MATERIALS & INTERFACES 2020; 12:14839-14854. [PMID: 32182035 DOI: 10.1021/acsami.9b22781] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A safe and efficient delivery system is critical for clinical application of siRNA. However, the conventional electrostatic interaction-based siRNA nanoplexes with bulk mixing preparation were always unsatisfactory for its stability and safety. In this study, the new core-shell lipid/PCL-PEI/siRNA nanoparticles (LPS NPs) endowing holonomic constraint of siRNA in the inner core were prepared by microfluidic technology. On the microfluidic chip, siRNAs were completely compressed into the inner hydrophilic core of reverse PCL-PEI micelles at a low N/P ratio of 5, followed by coating a neutral lipid membrane to form core-shell nanoparticles, which had a uniform size (120.2 ± 1.4 nm) and a negative charge (-8.8 ± 1.6 mV). Compared to bulk mixing-based LMS NPs, the lower usage of cationic PCL-PEI materials and stronger protection of siRNA in serum were found in the microfluidic-based LPS NPs. Furthermore, it was demonstrated that the LPS NPs exhibited significant downregulation of EGFR mRNA and protein expression level both in vitro and in vivo, and showed significant inhibition of tumor growth following systemic administration along with no obvious systemic toxicity. These findings demonstrated that the microfluidic-based lipid/polymer hybrid nanoassemblies would offer a promising siRNA delivery system for clinical application.
Collapse
Affiliation(s)
- Wei Wei
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Jing Sun
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Xi-Ying Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Xin Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Ru Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Chong Qiu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Hai-Tao Zhang
- School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Wen-Hao Pang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| |
Collapse
|
58
|
Huang HL, Lin WJ. Dual Peptide-Modified Nanoparticles Improve Combination Chemotherapy of Etoposide and siPIK3CA Against Drug-Resistant Small Cell Lung Carcinoma. Pharmaceutics 2020; 12:pharmaceutics12030254. [PMID: 32178266 PMCID: PMC7150975 DOI: 10.3390/pharmaceutics12030254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/18/2022] Open
Abstract
Small cell lung carcinoma (SCLC) is a highly aggressive form of malignancy with rapid recurrence and poor prognosis. The dual peptide-modified nanoparticles (NPs) for improving chemotherapy against drug-resistant small cell lung carcinoma cells has been developed. In this study, the SCLC targeting ligand, antagonist G peptide (AG), and cell-penetrating peptide, TAT, modified NPs were used to encapsulate both anticancer drugs etoposide (ETP) and PIK3CA small-interfering RNA (siPIK3CA). The ETP@NPs and siRNA@NPs had particle size 201.0 ± 1.9-206.5 ± 0.7 nm and 155.3 ± 12.4-169.1 ± 11.2 nm, respectively. The lyophilized ETP@NPs and siRNA@NPs maintained their particle size and zeta potential during 28-day storage without severe aggregation or dissociation. Either ETP@NPs or siRNA@NPs significantly reduced the IC50 of drugs by 2.5-5.5 folds and 2.4-3.9 folds, respectively, as compared to free ETP and siRNA/PEI nanocomplex in drug-resistant CD133(+) H69 cells. Herein, the IC50 of dual-peptide modified ETP@NPs and siRNA@NPs were prominently lower than single-peptide modified NPs. The synergistic effect (CI < 1) was further observed in co-treatment of ETP and siPIK3CA particularly delivered by dual-peptide modified NPs.
Collapse
Affiliation(s)
- Hsin-Lin Huang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan
| | - Wen Jen Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan
- Drug Research Center, College of Medicine, National Taiwan University, Taipei 10050, Taiwan
- Correspondence: ; Tel.: +886-2-33668765; Fax: +886-2-23919098
| |
Collapse
|
59
|
Li M, Li S, Zhou H, Tang X, Wu Y, Jiang W, Tian Z, Zhou X, Yang X, Wang Y. Chemotaxis-driven delivery of nano-pathogenoids for complete eradication of tumors post-phototherapy. Nat Commun 2020; 11:1126. [PMID: 32111847 PMCID: PMC7048836 DOI: 10.1038/s41467-020-14963-0] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery. The presence of several biological barriers impede the efficacy of nano-mediated drug delivery for solid cancer therapy. Here, the authors develop a nano-pathogenoid system that targets circulating neutrophils and show that it overcomes these biological barriers and improves tumour targeting and efficacy.
Collapse
Affiliation(s)
- Min Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China.,Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, Anhui, China.,College of Chemistry and Environmental Engineering, Shenzhen University, 518060, Shenzhen, Guangdong, China
| | - Shuya Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Han Zhou
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xinfeng Tang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Yi Wu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Wei Jiang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xuechang Zhou
- College of Chemistry and Environmental Engineering, Shenzhen University, 518060, Shenzhen, Guangdong, China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine and National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, 510006, Guangzhou, China
| | - Yucai Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China. .,Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, Anhui, China.
| |
Collapse
|
60
|
Lu T, Hu F, Yue H, Yang T, Ma G. The incorporation of cationic property and immunopotentiator in poly (lactic acid) microparticles promoted the immune response against chronic hepatitis B. J Control Release 2020; 321:576-588. [PMID: 32112853 DOI: 10.1016/j.jconrel.2020.02.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
Biodegradable microparticles (MPs) as vaccine adjuvants have sparked the passion of researchers in recent decades. However, it is still a huge challenge to develop an efficient vaccine delivery system to reverse chronic hepatitis B (CHB). Herein, we integrated a physiochemical merit and an immunopotentiator property in poly (lactic acid) (PLA) MPs and verified the therapeutic effect on CHB model mice. We prepared uniform MPs with insertion of cationic lipid didodecyldimethylammonium bromide (DDAB), which endowed a physiochemical merit for MPs. Such a DDAB-PLA (DP) group raised the recruitment of immune cells to the injection site along with the secretion of chemokines and pro-inflammatory cytokines, promoting the activation of antigen-presenting cells (APCs). Further combination of stimulator of interferon genes (STING) agonist 5,6-dimethylxanthenone-4-acetic acid (DMXAA) (DP-D) elevated 5.8-fold higher interferon regulatory factor 7 (IRF-7) expression compared to that for DP group. The DP group showed preferred lysosome escape advantage, which was in line with the DMXAA release behavior and the intracellular target of DMXAA. In addition, DP-D vaccine augmented the IFN-γ secreting splenocytes and motivated Th1-biased antibodies in a more efficient way than that for the DP group. In the CHB model, the MPs based vaccines achieved 50% HBsAg seroconversion rate, and HBcAg in the liver also got a reduction. DP-D produced higher amount of memory T/B cells to confer protection in a sustained manner. Present work thus provided a promising strategy, via integrating a fine-tuned physiochemical property and an immunopotentiator virtue in the MPs, which synergistically reinforced both humoral and cellular immune responses against CHB.
Collapse
Affiliation(s)
- Ting Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Fumin Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Tingyuan Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; Jiangsu National Synergetic Innovation Center for Advanced Materials, Nanjing 211816, PR China.
| |
Collapse
|
61
|
Oliveira ACN, Fernandes J, Gonçalves A, Gomes AC, Oliveira MECDR. Lipid-based Nanocarriers for siRNA Delivery: Challenges, Strategies and the Lessons Learned from the DODAX: MO Liposomal System. Curr Drug Targets 2020; 20:29-50. [PMID: 29968536 DOI: 10.2174/1389450119666180703145410] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/24/2018] [Accepted: 06/28/2018] [Indexed: 12/19/2022]
Abstract
The possibility of using the RNA interference (RNAi) mechanisms in gene therapy was one of the scientific breakthroughs of the last century. Despite the extraordinary therapeutic potential of this approach, the need for an efficient gene carrier is hampering the translation of the RNAi technology to the clinical setting. Although a diversity of nanocarriers has been described, liposomes continue to be one of the most attractive siRNA vehicles due to their relatively low toxicity, facilitated siRNA complexation, high transfection efficiency and enhanced pharmacokinetic properties. This review focuses on RNAi as a therapeutic approach, the challenges to its application, namely the nucleic acids' delivery process, and current strategies to improve therapeutic efficacy. Additionally, lipid-based nanocarriers are described, and lessons learned from the relation between biophysical properties and biological performance of the dioctadecyldimethylammonium:monoolein (DODAX: MO) system are explored. Liposomes show great potential as siRNA delivery systems, being safe nanocarriers to protect nucleic acids in circulation, extend their half-life time, target specific cells and reduce off-target effects. Nevertheless, several issues related to delivery must be overcome before RNAi therapies reach their full potential, namely target-cell specificity and endosomal escape. Understanding the relationship between biophysical properties and biological performance is an essential step in the gene therapy field.
Collapse
Affiliation(s)
- Ana C N Oliveira
- CBMA (Center of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal.,CFUM (Center of Physics), Department of Physics, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Joana Fernandes
- CBMA (Center of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Anabela Gonçalves
- CBMA (Center of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Andreia C Gomes
- CBMA (Center of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - M E C D Real Oliveira
- CFUM (Center of Physics), Department of Physics, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
62
|
Li L, Yang H, Chen P, Xin T, Zhou Q, Wei D, Zhang Y, Wang S. Trophoblast-Targeted Nanomedicine Modulates Placental sFLT1 for Preeclampsia Treatment. Front Bioeng Biotechnol 2020; 8:64. [PMID: 32117942 PMCID: PMC7026029 DOI: 10.3389/fbioe.2020.00064] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/27/2020] [Indexed: 12/28/2022] Open
Abstract
The overexpressed soluble fms-like tyrosine kinase 1 (sFLT-1) in placenta is considered to be a potential therapeutic target for preeclampsia (PE). How to achieve efficient intervention of sFLT1 expression in the placenta is an urgent problem to be solved. PEG-PLA nanoparticle generated by double-emulsion methods is a novel siRNA delivery system. Synthetic placental CSA binding peptide (P-CSA-BP) is effective for targeting lipid-polymer nanoparticle to the placenta. We conjugated P-CSA-BP to the surface of PEG-PLA nanoparticle to create a novel placenta specific sFLT1 siRNA delivery system for the therapy of PE. Nanoparticles were synthesized using double emulsion method and characterized by dynamic light scattering and transmission electron microscopy (TEM). RT-PCR was employed to evaluate mRNA level and protein level was analyzed by ELISA kit. The tissue distribution of nanoparticles was observed through ex vivo images. The concentrations of nanoparticles in organs were measured using high-performance liquid chromatography. T-NPsisFLT1 had higher efficiency than NPsisFLT1 in accumulating in HTR-8/SVneo cells and significantly decreased the expression of sFLT1. Intravenously administered T-NPsisFLT1 specifically accumulated in placentas of mice. sFLT1 mRNA level in placenta and protein level in serum were declined by T-NPsisFLT1. T-NPsisFLT1 shown no obvious toxic effect on both mother and fetus. The utility of T-NPsisFLT1 nanoparticles as a sFLT1 siRNA placenta specific delivery system significantly silenced sFLT1 in mice and is safe for both mother and fetus. This nanoparticle is a novel potential therapeutic strategy for PE.
Collapse
Affiliation(s)
- Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Key Laboratory of Birth Regulation and Control Technology of National Health and Family Planning Commission of China, Jinan, China
| | - Huijun Yang
- Key Laboratory of Birth Regulation and Control Technology of National Health and Family Planning Commission of China, Jinan, China.,Maternal and Child Health Care of Shandong Province, Jinan, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Qian Zhou
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dan Wei
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yanan Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shan Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
63
|
Wang Y, Luo YL, Chen YF, Lu ZD, Wang Y, Czarna A, Shen S, Xu CF, Wang J. Dually regulating the proliferation and the immune microenvironment of melanoma via nanoparticle-delivered siRNA targeting onco-immunologic CD155. Biomater Sci 2020; 8:6683-6694. [DOI: 10.1039/d0bm01420f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nanoparticle-delivered siRNA targeting CD155 for dual regulation of the proliferation and of the immune microenvironment of melanoma.
Collapse
Affiliation(s)
- Yan Wang
- Institutes for Life Sciences
- School of Medicine
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Ying-Li Luo
- Institutes for Life Sciences
- School of Medicine
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Yi-Fang Chen
- Institutes for Life Sciences
- School of Medicine
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Zi-Dong Lu
- Institutes for Life Sciences
- School of Medicine
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Yue Wang
- School of Biomedical Sciences and Engineering
- Guangzhou International Campus
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Anna Czarna
- Institutes for Life Sciences
- School of Medicine
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Song Shen
- School of Biomedical Sciences and Engineering
- Guangzhou International Campus
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering
- Guangzhou International Campus
- South China University of Technology
- Guangzhou 510006
- P.R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering
- Guangzhou International Campus
- South China University of Technology
- Guangzhou 510006
- P.R. China
| |
Collapse
|
64
|
Efficient nanocarriers of siRNA therapeutics for cancer treatment. Transl Res 2019; 214:62-91. [PMID: 31369717 DOI: 10.1016/j.trsl.2019.07.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/01/2019] [Accepted: 07/15/2019] [Indexed: 02/02/2023]
Abstract
Nanocarriers as drug delivery systems are promising and becoming popular, especially for cancer treatment. In addition to improving the pharmacokinetics of poorly soluble hydrophobic drugs by solubilizing them in a hydrophobic core, nanocarriers allow cancer-specific combination drug deliveries by inherent passive targeting phenomena and adoption of active targeting strategies. Nanoparticle-drug formulations can enhance the safety, pharmacokinetic profiles, and bioavailability of locally or systemically administered drugs, leading to improved therapeutic efficacy. Gene silencing by RNA interference (RNAi) is rapidly developing as a personalized field of cancer treatment. Small interfering RNAs (siRNAs) can be used to switch off specific cancer genes, in effect, "silence the gene, silence the cancer." siRNA can be used to silence specific genes that produce harmful or abnormal proteins. The activity of siRNA can be used to harness cellular machinery to destroy a corresponding sequence of mRNA that encodes a disease-causing protein. At present, the main barrier to implementing siRNA therapies in clinical practice is the lack of an effective delivery system that protects the siRNA from nuclease degradation, delivers to it to cancer cells, and releases it into the cytoplasm of targeted cancer cells, without creating adverse effects. This review provides an overview of various nanocarrier formulations in both research and clinical applications with a focus on combinations of siRNA and chemotherapeutic drug delivery systems for the treatment of multidrug resistant cancer. The use of various nanoparticles for siRNA-drug delivery, including liposomes, polymeric nanoparticles, dendrimers, inorganic nanoparticles, exosomes, and red blood cells for targeted drug delivery in cancer is discussed.
Collapse
|
65
|
Systematic chemical screening identifies disulfiram as a repurposed drug that enhances sensitivity to cisplatin in bladder cancer: a summary of preclinical studies. Br J Cancer 2019; 121:1027-1038. [PMID: 31673101 PMCID: PMC6964684 DOI: 10.1038/s41416-019-0609-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/24/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022] Open
Abstract
Background Since the standard gemcitabine and cisplatin (GC) chemotherapy for advanced bladder cancer yields limited therapeutic effect due to chemoresistance, it is a clinical challenge to enhance sensitivity to GC. Methods We performed high-throughput screening by using a library of known chemicals and repositionable drugs. A total of 2098 compounds were administered alone or with GC to human bladder cancer cells, and chemicals that enhanced GC effects were screened. Results Disulfiram (DSF), an anti-alcoholism drug, was identified as a candidate showing synergistic effects with cisplatin but not with gemcitabine in multiple cell lines. Co-administration of DSF with GC affected cellular localisation of a cisplatin efflux transporter ATP7A, increased DNA–platinum adducts and promoted apoptosis. Micellar DSF nanoparticles (DSF-NP) that stabilised DSF in vivo, enhanced the inhibitory effect of cisplatin in patient-derived and cell-based xenograft models without severe adverse effects. A drug susceptibility evaluation system by using cancer tissue-originated spheroid culture showed promise in identifying cases who would benefit from DSF with cisplatin. Conclusions The present study highlighted the advantage of drug repurposing to enhance the efficacy of anticancer chemotherapy. Repurposing of DSF to a chemotherapy sensitiser may provide additional efficacy with less expense by using an available drug with a well-characterised safety profile.
Collapse
|
66
|
Zhang Y, Shen S, Zhao G, Xu CF, Zhang HB, Luo YL, Cao ZT, Shi J, Zhao ZB, Lian ZX, Wang J. In situ repurposing of dendritic cells with CRISPR/Cas9-based nanomedicine to induce transplant tolerance. Biomaterials 2019; 217:119302. [PMID: 31271858 DOI: 10.1016/j.biomaterials.2019.119302] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 12/27/2022]
Abstract
Organ transplantation is the only effective method to treat end-stage organ failure. However, it is continuously plagued by immune rejection, which is mostly caused by T cell-mediated reactions. Dendritic cells (DCs) are professional antigen-presenting cells, and blocking the costimulatory signaling molecule CD40 in DCs inhibits T cell activation and induces transplant tolerance. In this study, to relieve graft rejection, Cas9 mRNA (mCas9) and a guide RNA targeting the costimulatory molecule CD40 (gCD40) were prepared and encapsulated into poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PEG-b-PLGA)-based cationic lipid-assisted nanoparticles (CLAN), denoted CLANmCas9/gCD40. CLAN effectively delivered mCas9/gCD40 into DCs and disrupted CD40 in DCs at the genomic level both in vitro and in vivo. After intravenous injection into an acute mouse skin transplant model, CLANmCas9/gCD40-mediated CD40 disruption significantly inhibited T cell activation, which reduced graft damage and prolonged graft survival. This work provides a promising strategy for reprogramming DCs with nanoparticles carrying the CRISPR/Cas9 system to abate transplant rejection.
Collapse
Affiliation(s)
- Yue Zhang
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Song Shen
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Gui Zhao
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Cong-Fei Xu
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China.
| | - Hou-Bing Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Ying-Li Luo
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhi-Ting Cao
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China
| | - Jia Shi
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhi-Bin Zhao
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China
| | - Jun Wang
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, PR China.
| |
Collapse
|
67
|
Baidoo SA, Sarkodie EK, Boakye-Yiadom KO, Kesse S. Nanomedicinal delivery systems for intelligent treatment of hepatocellular carcinoma. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
68
|
Ulkoski D, Bak A, Wilson JT, Krishnamurthy VR. Recent advances in polymeric materials for the delivery of RNA therapeutics. Expert Opin Drug Deliv 2019; 16:1149-1167. [PMID: 31498013 DOI: 10.1080/17425247.2019.1663822] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The delivery of nucleic acid therapeutics through non-viral carriers face multiple biological barriers that reduce their therapeutic efficiency. Despite great progress, there remains a significant technological gap that continues to limit clinical translation of these nanocarriers. A number of polymeric materials are being exploited to efficiently deliver nucleic acids and achieve therapeutic effects. Areas covered: We discuss the recent advances in the polymeric materials for the delivery of nucleic acid therapeutics. We examine the use of common polymer architectures and highlight the challenges that exist for their development from bench side to clinic. We also provide an overview of the most notable improvements made to circumvent such challenges, including structural modification and stimuli-responsive approaches, for safe and effective nucleic acid delivery. Expert opinion: It has become apparent that a universal carrier that follows 'one-size' fits all model cannot be expected for delivery of all nucleic acid therapeutics. Carriers need to be designed to exhibit sensitivity and specificity toward individual targets diseases/indications, and relevant subcellular compartments, each of which possess their own unique challenges. The ability to devise synthetic methods that control the molecular architecture enables the future development that allow for the construction of 'intelligent' designs.
Collapse
Affiliation(s)
- David Ulkoski
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Boston , USA
| | - Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Gothenburg , Sweden
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University , Nashville , TN , USA
| | | |
Collapse
|
69
|
Zhao G, Liu A, Zhang Y, Zuo ZQ, Cao ZT, Zhang HB, Xu CF, Wang J. Nanoparticle-delivered siRNA targeting Bruton's tyrosine kinase for rheumatoid arthritis therapy. Biomater Sci 2019; 7:4698-4707. [PMID: 31495833 DOI: 10.1039/c9bm01025d] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease that can cause irreversible joint deformity. There is still no cure for RA, and current therapeutics, including methotrexate and adalimumab, cause serious off-target effects and systemic immunosuppression, which in turn increases the risk of infection. Bruton's tyrosine kinase (BTK) in macrophages and B cells has been demonstrated to be a promising therapeutic target for RA. However, high doses of BTK inhibitors are required for efficient BTK suppression, which limits their clinical use. Small interfering RNA (siRNA) is promising for the silencing of specific genes and has been used for the treatment of multiple diseases. To deliver siRNA into macrophages and B cells for BTK gene silencing, we employed cationic lipid-assisted PEG-b-PLGA nanoparticles (CLANs) to encapsulate siRNA. We demonstrated that macrophages and B cells were able to efficiently ingest the CLANs both in vitro and in vivo. Thereafter, we encapsulated siRNA targeting BTK (siBTK) into the CLANs, denoted as CLANsiBTK, and demonstrated that CLANsiBTK significantly inhibited BTK expression in macrophages and B cells. In a collagen-induced mouse arthritis model, CLANsiBTK treatment dramatically reduced joint inflammation and other RA symptoms but showed no toxicity, proving that using CLANsiBTK is a promising approach for RA therapy.
Collapse
Affiliation(s)
- Gui Zhao
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, P. R. China
| | - An Liu
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, P. R. China
| | - Yue Zhang
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Zu-Qi Zuo
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, P. R. China
| | - Zhi-Ting Cao
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China. and National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| | - Hou-Bing Zhang
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, P. R. China
| | - Cong-Fei Xu
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China. and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jun Wang
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China. and Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China and Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, P. R. China
| |
Collapse
|
70
|
Huang H, Jiang CT, Shen S, Liu A, Gan YJ, Tong QS, Chen SB, Gao ZX, Du JZ, Cao J, Wang J. Nanoenabled Reversal of IDO1-Mediated Immunosuppression Synergizes with Immunogenic Chemotherapy for Improved Cancer Therapy. NANO LETTERS 2019; 19:5356-5365. [PMID: 31286779 DOI: 10.1021/acs.nanolett.9b01807] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Certain chemotherapeutics (e.g., oxaliplatin, OXA) can evoke effective antitumor immunity responses by inducing immunogenic cell death (ICD). Unfortunately, tumors always develop multiple immunosuppressive mechanisms, such as the upregulation of immunosuppressive factors, to counteract the effects of immunogenic chemotherapy. Indoleamine 2,3-dioxygenase-1 (IDO1), a tryptophan catabolic enzyme overexpressed in tumor-draining lymph nodes (TDLNs) and tumor tissues, plays a pivotal role in the generation of the immunosuppressive microenvironment. Reversing IDO1-mediated immunosuppression may strengthen the ICD-induced immune response. Herein, we developed a nanoenabled approach for IDO1 pathway interference, which is accomplished by delivering IDO1 siRNA to both TDLNs and tumor tissues with the help of cationic lipid-assisted nanoparticles (CLANs). We demonstrated that the contemporaneous administration of OXA and CLANsiIDO1 could achieve synergetic antitumor effects via promoting dendritic cell maturation, increasing tumor-infiltrating T lymphocytes and decreasing the number of regulatory T cells in a subcutaneous colorectal tumor model. We further proved that this therapeutic strategy is applicable for the treatment of orthotopic pancreatic tumors and offers a strong immunological memory effect, which can provide protection against tumor rechallenge.
Collapse
Affiliation(s)
| | | | | | - An Liu
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences , University of Science and Technology of China , Hefei 230027 , P.R. China
| | - Yun-Jiu Gan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences , University of Science and Technology of China , Hefei 230027 , P.R. China
| | | | | | | | | | | | - Jun Wang
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory , Guangzhou 510005 , P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education , South China University of Technology , Guangzhou 510006 , P.R. China
| |
Collapse
|
71
|
Abstract
The emergence of the CRISPR-Cas9 gene editing system has brought much hope and excitement to the field of gene therapy and the larger scientific community. However, in order for CRISPR-based therapies to be translated to the clinical setting, there is an urgent need to develop optimized vectors for their delivery. The delivery vector is a crucial determinant of the therapeutic efficacy of gene editing and should be designed to accommodate various factors including the form of the payload, the physiological environment, and the potential immune responses. Recently, biomaterials have become an attractive option for the delivery of Cas9 due to their tunability, biocompatibility and increasing efficacy at drug delivery. Biomaterials offer a unique solution for creating tailored vectors to meet the demands of various applications that cannot be easily met by other delivery methods. In this review, we will discuss the various biomaterial systems that have been used to deliver Cas9 in its plasmid, mRNA and protein forms. In addition, the functions of these materials will be reviewed to understand their roles in Cas9 delivery. Finally, the immune challenges associated with Cas9 and the delivery materials will be discussed as an understanding of the immune responses along with the functions of biomaterials will ultimately guide the field in designing new delivery systems for the clinical applications of CRISPR-Cas9.
Collapse
Affiliation(s)
- Joon Eoh
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, USA.
| | | |
Collapse
|
72
|
Gao X, Zou W, Jiang B, Xu D, Luo Y, Xiong J, Yan S, Wang Y, Tang Y, Chen C, Li H, Qiao H, Wang Q, Zou J. Experimental Study of Retention on the Combination of Bifidobacterium with High-Intensity Focused Ultrasound (HIFU) Synergistic Substance in Tumor Tissues. Sci Rep 2019; 9:6423. [PMID: 31015517 PMCID: PMC6478724 DOI: 10.1038/s41598-019-42832-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/09/2019] [Indexed: 01/04/2023] Open
Abstract
High intensity focused ultrasound (HIFU) has been recently regarded to be a new type of technique for non-invasive ablation of local tumors and HIFU synergists could significantly improve its therapeutic efficiency. The therapeutic efficiency of HIFU is greatly limited by the low retention of HIFU synergists in the target area and short residence time. This study aimed to explore a method to increase the deposition of HIFU synergists in tumors. Cationic lipid nanoparticle can be used to enhance the HIFU ablation effect, but there is still a problem for it that the deposition amount in the tumor tissue is small and the residence time is short. Bifidobacterium is highly biosafe and can be selectively colonized in the hypoxic zone of tumor tissue. Cationic lipid nanoparticles can be observed in vitro by attachment to bifidobacterium by electrostatic adsorption. And the effect of the proliferation of bifidobacterium in tumor tissues on the retention amount and retention time of cationic lipid nanoparticles in vivo was evaluated. Results showed that the cationic lipid nanoparticles were linked to the surface of Bifidobacterium effectively in vitro, while in vivo, the retention amount and retention time of cationic lipid nanoparticles could be increased by Bifidobacterium in tumor tissues, which provided a new method for improving the therapeutic efficiency of HIFU.
Collapse
Affiliation(s)
- Xuan Gao
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Wenjuan Zou
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Binglei Jiang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Die Xu
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yong Luo
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Xiong
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Sijing Yan
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Yaotai Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yu Tang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Chun Chen
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Huanan Li
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Hai Qiao
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Qi Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jianzhong Zou
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
73
|
Shi J, Liu S, Yu Y, He C, Tan L, Shen YM. RGD peptide-decorated micelles assembled from polymer-paclitaxel conjugates towards gastric cancer therapy. Colloids Surf B Biointerfaces 2019; 180:58-67. [PMID: 31028965 DOI: 10.1016/j.colsurfb.2019.04.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/09/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022]
Abstract
Development of polymer-drug conjugate capable of controlled drug release is urgently needed for gastric cancer therapy. Herein, arginine-glycine-aspartic acid (RGD)-decorated polyethylene glycol (PEG)-paclitaxel (PTX) conjugates containing disulfide linkage were synthesized. The amphiphilic PEG-PTX conjugates were found to assemble into micelles (RGD@Micelles), which would be decomposed under the reduction of glutathione (GSH) and finally release PTX in weakly acidic conditions characteristic of intracellular environment. The RGD@Micelles were spherical nanoparticles with an average hydrodynamic size of ˜50 nm, which were stable in physiological environment. The release of PTX from the micelles in response to GSH was investigated. In vitro cell assay suggested that the RGD@Micelles could target the gastric cancer cells and inhibit cell proliferation by inducing apoptosis. In vivo experiments indicated that the RGD@Micelles could be delivered to the tumor site and inhibit the tumor growth efficiently by releasing PTX inside the tumor cells. This type of micelles exhibited high therapeutic efficacy and low side effects, providing new insights into targeted drug delivery for gastric cancer therapy.
Collapse
Affiliation(s)
- Jingwen Shi
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuiping Liu
- College of Textile and Clothing, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Yuan Yu
- College of Textile & Clothing, Jiangnan University, Wuxi, 214122, China
| | - Changyu He
- Shanghai Key Laboratory of Gastric Neoplasms, Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lianjiang Tan
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yu-Mei Shen
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
74
|
Kumari P, Rompicharla SVK, Bhatt H, Ghosh B, Biswas S. Development of chlorin e6-conjugated poly(ethylene glycol)-poly(d,l-lactide) nanoparticles for photodynamic therapy. Nanomedicine (Lond) 2019; 14:819-834. [DOI: 10.2217/nnm-2018-0255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: In this study, we developed a chlorin e6-conjugated methoxy-poly(ethylene glycol)-poly(d,l-lactide) (mPEG-PLA-Ce6) amphiphilic polymer, which self-assembled to form stable nanoparticles. Materials & methods: The nanoparticles were characterized for particle size, ζ-potential and singlet oxygen (1O2) generation. Cellular internalization and phototoxicity were investigated against monolayer and 3D spheroids of human lung adenocarcinoma cells (A549). Results & conclusion: mPEG-PLA-Ce6 exhibited a size of 149.72 ± 3.51 nm and ζ-potential of -24.82 ± 2.94 mV. The 1O2 generation by mPEG-PLA-Ce6 in water was considerably higher than free chlorin e6. The nanoparticles showed enhanced cellular internalization and phototoxicity in monolayer and 3D spheroids. The developed mPEG-PLA-Ce6 has potential application as a nanocarrier of chlorin e6 for photodynamic therapy of solid tumors.
Collapse
Affiliation(s)
- Preeti Kumari
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Sri Vishnu Kiran Rompicharla
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Himanshu Bhatt
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| |
Collapse
|
75
|
Dong Y, Siegwart DJ, Anderson DG. Strategies, design, and chemistry in siRNA delivery systems. Adv Drug Deliv Rev 2019; 144:133-147. [PMID: 31102606 DOI: 10.1016/j.addr.2019.05.004] [Citation(s) in RCA: 383] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/03/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022]
Abstract
Emerging therapeutics that utilize RNA interference (RNAi) have the potential to treat broad classes of diseases due to their ability to reversibly silence target genes. In August 2018, the FDA approved the first siRNA therapeutic, called ONPATTRO™ (Patisiran), for the treatment of transthyretin-mediated amyloidosis. This was an important milestone for the field of siRNA delivery that opens the door for additional siRNA drugs. Currently, >20 small interfering RNA (siRNA)-based therapies are in clinical trials for a wide variety of diseases including cancers, genetic disorders, and viral infections. To maximize therapeutic benefits of siRNA-based drugs, a number of chemical strategies have been applied to address issues associated with efficacy, specificity, and safety. This review focuses on the chemical perspectives behind non-viral siRNA delivery systems, including siRNA synthesis, siRNA conjugates, and nanoparticle delivery using nucleotides, lipids, and polymers. Tracing and understanding the chemical development of strategies to make siRNAs into drugs is important to guide development of additional clinical candidates and enable prolonged success of siRNA therapeutics.
Collapse
Affiliation(s)
- Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| | - Daniel G Anderson
- Deparment of Chemical Engineering, David H. Koch Institute for Integrative Cancer Research, Department of Chemistry, Institute for Medical Engineering and Science, and Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, United States.
| |
Collapse
|
76
|
Xu CF, Iqbal S, Shen S, Luo YL, Yang X, Wang J. Development of "CLAN" Nanomedicine for Nucleic Acid Therapeutics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900055. [PMID: 30884095 DOI: 10.1002/smll.201900055] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/19/2019] [Indexed: 05/17/2023]
Abstract
Nucleic acid-based macromolecules have paved new avenues for the development of therapeutic interventions against a spectrum of diseases; however, their clinical translation is limited by successful delivery to the target site and cells. Therefore, numerous systems have been developed to overcome delivery challenges to nucleic acids. From the viewpoint of clinical translation, it is highly desirable to develop systems with clinically validated materials and controllability in synthesis. With this in mind, a cationic lipid assisted PEG-b-PLA nanoparticle (CLAN) is designed that is capable of protecting nucleic acids via encapsulation inside the aqueous core, and delivers them to target cells, while maintaining or improving nucleic acid function. The system is formulated from clinically validated components (PEG-b-PLA and its derivatives) and can be scaled-up for large scale manufacturing, offering potential for its future use in clinical applications. Here, the development and working mechanisms of CLANs, the ways to improve its delivery efficacy, and its application in various disease treatments are summarized. Finally, a prospective for the further development of CLAN is also discussed.
Collapse
Affiliation(s)
- Cong-Fei Xu
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangdong, 510006, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Shoaib Iqbal
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Song Shen
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangdong, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Ying-Li Luo
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangdong, 510006, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, P. R. China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangdong, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Jun Wang
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangdong, 510006, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| |
Collapse
|
77
|
Ning Q, Liu YF, Ye PJ, Gao P, Li ZP, Tang SY, He DX, Tang SS, Wei H, Yu CY. Delivery of Liver-Specific miRNA-122 Using a Targeted Macromolecular Prodrug toward Synergistic Therapy for Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:10578-10588. [PMID: 30802029 DOI: 10.1021/acsami.9b00634] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Hepatocellular carcinoma (HCC) poses a great threat to human health. The elegant combination of gene therapy and chemotherapy by nanocarriers has been repeatedly highlighted to realize enhanced therapeutic efficacy relative to monotreatment. However, the leading strategy to achieve the efficient codelivery of the gene and drug remains the electrostatic condensation with the nucleic acid and the hydrophobic encapsulation of drug molecules by the nanocarriers, which suffers substantially from premature drug leakage during circulation and severe off-target-associated side effects. To address these issues, we reported in this study the codelivery of liver-specific miRNA-122 and anti-cancer drug 5-fluorouracil (5-Fu) using a macromolecular prodrug approach, that is, electrostatic condensation with miRNA-122 using galactosylated-chitosan-5-fluorouracil (GC-FU). The delivery efficacy was evaluated comprehensively in vitro and in vivo. Specifically, the biocompatibility of GC-FU/miR-122 nanoparticles (NPs) was assessed by hemolysis activity analysis, BSA adsorption test, and cell viability assay in both normal liver cells (L02 cells) and endothelial cells. The resulting codelivery systems showed enhanced blood and salt stability, efficient proliferation inhibition of HCC cells, and further induction apoptosis of HCC cells, as well as downregulated expression of ADAM17 and Bcl-2. The strategy developed herein is thus a highly promising platform for an effective codelivery of miRNA-122 and 5-Fu with facile fabrication and great potential for the clinical translation toward HCC synergistic therapy.
Collapse
Affiliation(s)
- Qian Ning
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System , Hunan University of Medicine , Huaihua 418000 , China
| | | | | | - Pei Gao
- Chemistry Department , Eastern Kentucky University , Richmond , Kentucky 40475 , United States
| | | | | | | | - Sheng-Song Tang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System , Hunan University of Medicine , Huaihua 418000 , China
| | - Hua Wei
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System , Hunan University of Medicine , Huaihua 418000 , China
| | - Cui-Yun Yu
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System , Hunan University of Medicine , Huaihua 418000 , China
| |
Collapse
|
78
|
Jiang Q, Chen X, Liang H, Nie Y, Jin R, Barz M, Yue D, Gu Z. Multistage rocket: integrational design of a prodrug-based siRNA delivery system with sequential release for enhanced antitumor efficacy. NANOSCALE ADVANCES 2019; 1:498-507. [PMID: 36132232 PMCID: PMC9473180 DOI: 10.1039/c8na00191j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/12/2018] [Indexed: 05/24/2023]
Abstract
An integrated peptide-camptothecin prodrug (RSC) system was designed as a nano-sized multistage rocket for the efficient complexation and controlled sequential release of siRNA and anticancer drug under tumor-relevant reductive and esterase-enriched conditions, which facilitated the avoidance of negative interactions and maximized the synergistic effect.
Collapse
Affiliation(s)
- Qian Jiang
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610064 P. R. China
- Department of Pharmacy, The Second People's Hospital of Chengdu Chengdu 610017 P. R. China
| | - Xiaobing Chen
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610064 P. R. China
| | - Hong Liang
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610064 P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610064 P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610064 P. R. China
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz Duesbergweg 10-14 55099 Mainz Germany
| | - Dong Yue
- National Engineering Research Center for Biomaterials, Sichuan University Chengdu 610064 P. R. China
| | - Zhongwei Gu
- College of Materials Science and Engineering, Nanjing Tech University 30 South Puzhu Road Nanjing 211816 P. R. China
| |
Collapse
|
79
|
Polyester based nanovehicles for siRNA delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1006-1015. [DOI: 10.1016/j.msec.2018.05.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/12/2018] [Accepted: 05/07/2018] [Indexed: 12/18/2022]
|
80
|
Polyester-based nanoparticles for nucleic acid delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:983-994. [DOI: 10.1016/j.msec.2018.07.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
|
81
|
Park G, Son B, Kang J, Lee S, Jeon J, Kim JH, Yi GR, Youn H, Moon C, Nam SY, Youn B. LDR-Induced miR-30a and miR-30b Target the PAI-1 Pathway to Control Adverse Effects of NSCLC Radiotherapy. Mol Ther 2018; 27:342-354. [PMID: 30424954 DOI: 10.1016/j.ymthe.2018.10.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy has been a central part in curing non-small cell lung cancer (NSCLC). However, it is possible that not all of the tumor cells are destroyed by radiation; therefore, it is important to effectively control residual tumor cells that could become aggressive and resistant to radiotherapy. In this study, we aimed to investigate the molecular mechanism of decreased NSCLC radioresistance by low-dose radiation (LDR) pretreatment. The results indicated that miR-30a and miR-30b, which effectively inhibited plasminogen activator inhibitor-1 (PAI-1), were overexpressed by treatment of LDR to NSCLC cells. Phosphorylation of Akt and ERK, the downstream survival signals of PAI-1, was decreased by PAI-1 inhibition. Reduced cell survival and epithelial-mesenchymal transition by PAI-1 inhibition were confirmed in NSCLC cells. Moreover, in vivo orthotopic xenograft mouse models with 7C1 nanoparticles to deliver miRNAs showed that tumor growth and aggressiveness were efficiently decreased by LDR treatment followed by radiotherapy. Taken together, the present study suggested that PAI-1, whose expression is regulated by LDR, was critical for controlling surviving tumor cells after radiotherapy.
Collapse
Affiliation(s)
- Gaeul Park
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - JiHoon Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea; Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Republic of Korea
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Jaewan Jeon
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea; Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Republic of Korea
| | - Joo-Hyung Kim
- Department of Chemistry, Molecular Design Institute, New York University, New York, NY 10003, USA
| | - Gi-Ra Yi
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seon Young Nam
- Low-Dose Radiation Research Team, Radiation Health Institute, Korea Hydro & Nuclear Power Co., Ltd., Seoul 01450, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea; Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
82
|
Liu Y, Zhao G, Xu CF, Luo YL, Lu ZD, Wang J. Systemic delivery of CRISPR/Cas9 with PEG-PLGA nanoparticles for chronic myeloid leukemia targeted therapy. Biomater Sci 2018; 6:1592-1603. [PMID: 29725684 DOI: 10.1039/c8bm00263k] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Chronic myeloid leukemia (CML), which is characterized by the Philadelphia translocation, which fuses breakpoint cluster region (BCR) sequences from chromosome 22 upstream of the Abelson murine leukemia viral oncogene homolog (ABL) on chromosome 9, requires specific and efficient treatment. The CRISPR/Cas9 system, with its mechanism of specific DNA complementary recognition by engineered guide RNA (gRNA), allows the development of novel therapeutics for CML. To achieve targeted therapy of CML with the CRISPR/Cas9 system, we encapsulated a CRISPR/Cas9 plasmid (pCas9) expressing gRNA targeting the overhanging fusion region of the BCR-ABL gene (pCas9/gBCR-ABL) with poly(ethylene glycol)-b-poly(lactic acid-co-glycolic acid) (PEG-PLGA)-based cationic lipid-assisted polymeric nanoparticles (CLANs), which specifically disrupted the CML-related BCR-ABL gene while sparing the BCR and ABL genes in normal cells. After intravenous injection, CLANs carrying pCas9/gBCR-ABL (CLANpCas9/gBCR-ABL) efficiently knocked out the BCR-ABL fusion gene of CML cells and improved the survival of a CML mouse model, indicating that the combination of the CRISPR/Cas9 system with nanocarriers is a promising strategy for targeted treatment of CML.
Collapse
Affiliation(s)
- Yang Liu
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | | | | | | | | | | |
Collapse
|
83
|
Targeting of NLRP3 inflammasome with gene editing for the amelioration of inflammatory diseases. Nat Commun 2018; 9:4092. [PMID: 30291237 PMCID: PMC6173702 DOI: 10.1038/s41467-018-06522-5] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 09/04/2018] [Indexed: 12/25/2022] Open
Abstract
The NLRP3 inflammasome is a well-studied target for the treatment of multiple inflammatory diseases, but how to promote the current therapeutics remains a large challenge. CRISPR/Cas9, as a gene editing tool, allows for direct ablation of NLRP3 at the genomic level. In this study, we screen an optimized cationic lipid-assisted nanoparticle (CLAN) to deliver Cas9 mRNA (mCas9) and guide RNA (gRNA) into macrophages. By using CLAN encapsulating mCas9 and gRNA-targeting NLRP3 (gNLRP3) (CLANmCas9/gNLRP3), we disrupt NLRP3 of macrophages, inhibiting the activation of the NLRP3 inflammasome in response to diverse stimuli. After intravenous injection, CLANmCas9/gNLRP3 mitigates acute inflammation of LPS-induced septic shock and monosodium urate crystal (MSU)-induced peritonitis. In addition, CLANmCas9/gNLRP3 treatment improves insulin sensitivity and reduces adipose inflammation of high-fat-diet (HFD)-induced type 2 diabetes (T2D). Thus, our study provides a promising strategy for treating NLRP3-dependent inflammatory diseases and provides a carrier for delivering CRISPR/Cas9 into macrophages. Activation of the NLRP3 inflammasome triggers the production of inflammatory cytokines. Here, the authors inactivate NLRP3 in macrophages using CRISPR/Cas9 encapsulated in nanoparticles, and show that administration in mice is effective in preventing septic shock and peritonitis, and in improving diabetes-associated inflammation and insulin resistance.
Collapse
|
84
|
Fan YN, Li M, Luo YL, Chen Q, Wang L, Zhang HB, Shen S, Gu Z, Wang J. Cationic lipid-assisted nanoparticles for delivery of mRNA cancer vaccine. Biomater Sci 2018; 6:3009-3018. [PMID: 30264063 DOI: 10.1039/c8bm00908b] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Message RNA-based vaccines with prominent advantages such as facile production, no requirement for nuclear entry and high safety without the need for integration into host genome have been shown to be potent activators of the cytotoxic immune system. However, wider applications of mRNA-based therapeutics have been hindered because of their intrinsically high vulnerability to expressed nucleases and difficulty while entering antigen-presenting cells (APCs) directly. Here, we investigated the potential of cationic lipid-assisted nanoparticles (CLAN), which form a clinically translatable nucleic acid delivery system working as a carrier of an mRNA vaccine. We found that CLAN encapsulating mRNA encoding antigen could effectively stimulate the maturation of dendritic cells (DCs) and promote the activation and proliferation of antigen-specific T cells both in vitro and in vivo. Intravenous immunization of mice with CLAN containing mRNA encoding ovalbumin (OVA) provoked a strong OVA-specific T-cell response and slowed tumor growth in an aggressive E·G7-OVA lymphoma model. Collectively, CLAN proved to be a promising platform for mRNA vaccine delivery.
Collapse
Affiliation(s)
- Ya-Nan Fan
- School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, P. R. China
| | - Min Li
- School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, P. R. China
| | - Ying-Li Luo
- School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, P. R. China
| | - Qian Chen
- Department of Bioengineering, California Nanosystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Li Wang
- School of Life Sciences, University of Science & Technology of China, Hefei, Anhui 230027, P. R. China
| | - Hou-Bing Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Song Shen
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China. and National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China and Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zhen Gu
- Department of Bioengineering, California Nanosystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jun Wang
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China. and National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| |
Collapse
|
85
|
Inhibition of miR-449a Promotes Cartilage Regeneration and Prevents Progression of Osteoarthritis in In Vivo Rat Models. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:322-333. [PMID: 30326428 PMCID: PMC6197768 DOI: 10.1016/j.omtn.2018.09.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 09/21/2018] [Accepted: 09/23/2018] [Indexed: 12/20/2022]
Abstract
Traumatic and degenerative lesions of articular cartilage usually progress to osteoarthritis (OA), a leading cause of disability in humans. MicroRNAs (miRNAs) can regulate the differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) and play important roles in the expression of genes related to OA. However, their functional roles in OA remain poorly understood. Here, we have examined miR-449a, which targets sirtuin 1 (SIRT1) and lymphoid enhancer-binding factor-1 (LEF-1), and observed its effects on damaged cartilage. The levels of chondrogenic markers and miR-449a target genes increased during chondrogenesis in anti-miR-449a-transfected hBMSCs. A locked nucleic acid (LNA)-anti-miR-449a increased cartilage regeneration and expression of type II collagen and aggrecan on the regenerated cartilage surface in acute defect and OA models. Furthermore, intra-articular injection of LNA-anti-miR-449a prevented disease progression in the OA model. Our study indicates that miR-449a may be a novel potential therapeutic target for age-related joint diseases like OA.
Collapse
|
86
|
Exploring the role of polymeric conjugates toward anti-cancer drug delivery: Current trends and future projections. Int J Pharm 2018; 548:500-514. [DOI: 10.1016/j.ijpharm.2018.06.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
|
87
|
Xiao C, Wang F, Hou J, Zhu X, Luo Y, Xiong JW. Nanoparticle-mediated siRNA Gene-silencing in Adult Zebrafish Heart. J Vis Exp 2018. [PMID: 30102293 DOI: 10.3791/58054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mammals have a very limited capacity to regenerate the heart after myocardial infarction. On the other hand, the adult zebrafish regenerates its heart after apex resection or cryoinjury, making it an important model organism for heart regeneration study. However, the lack of loss-of-function methods for adult organs has restricted insights into the mechanisms underlying heart regeneration. RNA interference via different delivery systems is a powerful tool for silencing genes in mammalian cells and model organisms. We have previously reported that siRNA-encapsulated nanoparticles successfully enter cells and result in a remarkable gene-specific knockdown in the regenerating adult zebrafish heart. Here, we present a simple, rapid, and efficient protocol for the dendrimer-mediated siRNA delivery and gene-silencing in the regenerating adult zebrafish heart. This method provides an alternative approach for determining gene functions in adult organs in zebrafish and can be extended to other model organisms as well.
Collapse
Affiliation(s)
- Chenglu Xiao
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University
| | - Fang Wang
- Department of Biomedical Engineering, College of Engineering, Peking University
| | - Junjie Hou
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University;
| |
Collapse
|
88
|
Hirano T, Nakatani A, Kawamata Y. Selective Synthesis of Monoadduct Derivatives of Triaminopentanoic Acid Anhydride for Time-resolved Fluorescence-mediated Polymer Concentration Measurement. CHEM LETT 2018. [DOI: 10.1246/cl.180036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Takehiro Hirano
- Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Atsushi Nakatani
- Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Yuji Kawamata
- Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
89
|
Liu Y, Cao ZT, Xu CF, Lu ZD, Luo YL, Wang J. Optimization of lipid-assisted nanoparticle for disturbing neutrophils-related inflammation. Biomaterials 2018; 172:92-104. [PMID: 29723758 DOI: 10.1016/j.biomaterials.2018.04.052] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/22/2018] [Accepted: 04/25/2018] [Indexed: 12/31/2022]
Abstract
Inflammation is closely related to the development of many diseases and is commonly characterized by abnormal infiltration of immune cells, especially neutrophils. The current therapeutics of inflammatory diseases give little attention to direct modulation of these diseases with respect to immune cells. Nanoparticles are applied for efficient drug delivery into the disease-related immune cells, but their performance is significantly affected by their surface properties. In this study, to optimize the properties of nanoparticles for modulating neutrophils-related inflammation, we prepared a library of poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-b-PLGA)-based cationic lipid-assisted nanoparticles (CLANs) with different surface PEG density and surface charge. Optimized CLANs for neutrophils targeting were screened in high-fat diet (HFD)-induced type 2 diabetes (T2D) mice. Then, a CRISPR-Cas9 plasmid expressing a guide RNA (gRNA) targeting neutrophil elastase (NE) was encapsulated into the optimized CLAN and denoted as CLANpCas9/gNE. After intravenous injection, CLANpCas9/gNE successfully disrupted the NE gene of neutrophils and mitigated the insulin resistance of T2D mice via reducing the inflammation in epididymal white adipose tissue (eWAT) and in the liver. This strategy provides an example of abating the inflammatory microenvironment by directly modulating immune cells with nanoparticles carrying genome editing tools.
Collapse
Affiliation(s)
- Yang Liu
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Zhi-Ting Cao
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Cong-Fei Xu
- Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China.
| | - Zi-Dong Lu
- Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China
| | - Ying-Li Luo
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Jun Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China.
| |
Collapse
|
90
|
Luo YL, Xu CF, Li HJ, Cao ZT, Liu J, Wang JL, Du XJ, Yang XZ, Gu Z, Wang J. Macrophage-Specific in Vivo Gene Editing Using Cationic Lipid-Assisted Polymeric Nanoparticles. ACS NANO 2018; 12:994-1005. [PMID: 29314827 DOI: 10.1021/acsnano.7b07874] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The CRISPR/Cas9 gene editing technology holds promise for the treatment of multiple diseases. However, the inability to perform specific gene editing in targeted tissues and cells, which may cause off-target effects, is one of the critical bottlenecks for therapeutic application of CRISPR/Cas9. Herein, macrophage-specific promoter-driven Cas9 expression plasmids (pM458 and pM330) were constructed and encapsulated in cationic lipid-assisted PEG-b-PLGA nanoparticles (CLAN). The obtained nanoparticles encapsulating the CRISPR/Cas9 plasmids were able to specifically express Cas9 in macrophages as well as their precursor monocytes both in vitro and in vivo. More importantly, after further encoding a guide RNA targeting Ntn1 (sgNtn1) into the plasmid, the resultant CLANpM330/sgNtn1 successfully disrupted the Ntn1 gene in macrophages and their precursor monocytes in vivo, which reduced expression of netrin-1 (encoded by Ntn1) and subsequently improved type 2 diabetes (T2D) symptoms. Meanwhile, the Ntn1 gene was not disrupted in other cells due to specific expression of Cas9 by the CD68 promoter. This strategy provides alternative avenues for specific in vivo gene editing with the CRISPR/Cas9 system.
Collapse
Affiliation(s)
- Ying-Li Luo
- School of Life Sciences, University of Science and Technology of China , Hefei, Anhui 230027, People's Republic of China
| | - Cong-Fei Xu
- Institutes for Life Sciences and School of Medicine, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
| | - Hong-Jun Li
- Institutes for Life Sciences and School of Medicine, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
| | - Zhi-Ting Cao
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China , Hefei, Anhui 230027, People's Republic of China
| | - Jing Liu
- School of Life Sciences, University of Science and Technology of China , Hefei, Anhui 230027, People's Republic of China
| | - Ji-Long Wang
- School of Life Sciences, University of Science and Technology of China , Hefei, Anhui 230027, People's Republic of China
| | - Xiao-Jiao Du
- Institutes for Life Sciences and School of Medicine, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
| | - Xian-Zhu Yang
- Institutes for Life Sciences and School of Medicine, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Jun Wang
- School of Life Sciences, University of Science and Technology of China , Hefei, Anhui 230027, People's Republic of China
- Institutes for Life Sciences and School of Medicine, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology , Guangzhou, Guangdong 510006, People's Republic of China
- Research Institute for Food Nutrition and Human Health , Guangzhou, Guangdong 510006, People's Republic of China
| |
Collapse
|
91
|
Wang Y, Li SY, Shen S, Wang J. Protecting neurons from cerebral ischemia/reperfusion injury via nanoparticle-mediated delivery of an siRNA to inhibit microglial neurotoxicity. Biomaterials 2018; 161:95-105. [PMID: 29421566 DOI: 10.1016/j.biomaterials.2018.01.039] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/18/2018] [Accepted: 01/25/2018] [Indexed: 01/14/2023]
Abstract
Complement component C3 (C3) plays a central role in microglial neurotoxicity following cerebral ischemia/reperfusion (I/R) injury. In this study, we focused on the role of nanoparticles loaded with C3 siRNA (NPsiC3) in inhibiting microglial neurotoxicity after brain (I/R) injury. NPsiC3 inhibited the hypoxia/re-oxygenation-induced increase in C3 expression in microglia in vitro. Importantly, treatment with NPsiC3 decreased C3b deposition on neurons and reduced microglia-mediated neuronal damage under hypoxia/re-oxygen conditions. Nanoparticles could effectively deliver C3-siRNA from the blood into ischemic penumbra across the blood-brain barrier (BBB) and significantly decrease C3 expression in microglia and ischemic brain tissue, while reducing the number of infiltrating inflammatory cells and the concentration of pro-inflammatory factors in the penumbra. Furthermore, NPsiC3 also prevented neuronal apoptosis, reduced the volume of the ischemic zone, and substantially improved functional recovery after I/R injury. Therefore, the NPsiC3-induced inhibition of microglial neurotoxicity represents a novel therapeutic strategy for treating brain I/R injury.
Collapse
Affiliation(s)
- Ye Wang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Shi-Yong Li
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China; Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| | - Song Shen
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong 510006, PR China
| | - Jun Wang
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong 510006, PR China; Research Institute for Food Nutrition and Human Health, Guangzhou, Guangdong 510006, PR China.
| |
Collapse
|
92
|
Shen S, Zhang Y, Chen KG, Luo YL, Wang J. Cationic Polymeric Nanoparticle Delivering CCR2 siRNA to Inflammatory Monocytes for Tumor Microenvironment Modification and Cancer Therapy. Mol Pharm 2018; 15:3642-3653. [PMID: 29337566 DOI: 10.1021/acs.molpharmaceut.7b00997] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Accumulating evidence has confirmed that malignant tumors have a complex microenvironment, which consists of a heterogeneous collection of tumor cells and other cell subsets (including the full gamut of immune cells). Tumor-associated macrophages (TAMs), derived from circulating Ly6Chi monocytes, constitute the most substantial fraction of tumor-infiltrating immune cells in nearly all cancer types and contribute to tumor progression, vascularization, metastasis, immunosuppression, and therapeutic resistance. Interrupting monocyte recruitment to tumor tissues by disturbing pivotal signaling pathways (such as CCL2-CCR2) is viewed as one of the most promising avenues for tumor microenvironment manipulation and cancer therapy. One critical issue for monocyte-based therapy is to deliver therapeutic agents into monocytes efficiently. In the present study, we systematically investigated the relationship between the surface potential and the biodistribution of polymeric nanoparticles in monocytes in vivo, aiming to screen and identify an appropriate delivery system for monocyte targeting, and we found that cationic nanoparticles have a higher propensity to accumulate in monocytes compared with their neutral counterparts. We further demonstrated that siCCR2-encapsulated cationic nanoparticle (CNP/siCCR2) could modify immunosuppressive tumor microenvironment more efficiently and exhibit superior antitumor effect in an orthotopic murine breast cancer model.
Collapse
Affiliation(s)
- Song Shen
- Institutes for Life Sciences and School of Medicine , South China University of Technology , Guangzhou , Guandong 510006 , P. R. China.,National Engineering Research Center for Tissue Restoration and Reconstruction , Guangzhou , Guangdong 510006 , P. R. China
| | - Yue Zhang
- School of Life Sciences , University of Science & Technology of China , Hefei , Anhui 230027 , P. R. China
| | - Kai-Ge Chen
- School of Life Sciences , University of Science & Technology of China , Hefei , Anhui 230027 , P. R. China
| | - Ying-Li Luo
- School of Life Sciences , University of Science & Technology of China , Hefei , Anhui 230027 , P. R. China
| | - Jun Wang
- School of Life Sciences , University of Science & Technology of China , Hefei , Anhui 230027 , P. R. China.,Institutes for Life Sciences and School of Medicine , South China University of Technology , Guangzhou , Guandong 510006 , P. R. China.,National Engineering Research Center for Tissue Restoration and Reconstruction , Guangzhou , Guangdong 510006 , P. R. China.,Research Institute for Food Nutrition and Human Health , South China University of Technology , Guangzhou 510641 , P. R. China
| |
Collapse
|
93
|
EV, Microvesicles/MicroRNAs and Stem Cells in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:123-135. [PMID: 29754178 DOI: 10.1007/978-3-319-74470-4_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The role of extracellular vesicles (EV) in carcinogenesis has become the focus of much research. These microscopic messengers have been found to regulate immune system function, particularly in tumorigenesis, as well as conditioning future metastatic sites for the attachment and growth of tumor tissue. Through an interaction with a range of host tissues, EVs are able to generate a pro-tumor environment that is essential for tumorigenesis. These small nanovesicles are an ideal candidate for a non-invasive indicator of pathogenesis and/or disease progression as they can display individualized nucleic acid, protein, and lipid expression profiles that are often reflective of disease state, and can be easily detected in bodily fluids, even after extended cryo-storage. Furthermore, the ability of EVs to securely transport signaling molecules and localize to distant tissues suggests these particles may greatly improve the delivery of therapeutic treatments, particularly in cancer. In this chapter, we discuss the role of EV in the identification of new diagnostic and prognostic cancer biomarkers, as well as the development of novel EV-based cancer therapies.
Collapse
|
94
|
Du XJ, Wang JL, Iqbal S, Li HJ, Cao ZT, Wang YC, Du JZ, Wang J. The effect of surface charge on oral absorption of polymeric nanoparticles. Biomater Sci 2018; 6:642-650. [DOI: 10.1039/c7bm01096f] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Positively charged nanoparticles showed a favorable distribution in the small intestine, and significantly improved oral bioavailability.
Collapse
Affiliation(s)
- Xiao-Jiao Du
- Institutes for Life Sciences and School of Medicine
- South China University of Technology
- Guangzhou
- China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| | - Ji-Long Wang
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- China
| | - Shoaib Iqbal
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- China
| | - Hong-Jun Li
- Institutes for Life Sciences and School of Medicine
- South China University of Technology
- Guangzhou
- China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| | - Zhi-Ting Cao
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- China
| | - Yu-Cai Wang
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- China
| | - Jin-Zhi Du
- Institutes for Life Sciences and School of Medicine
- South China University of Technology
- Guangzhou
- China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| | - Jun Wang
- Institutes for Life Sciences and School of Medicine
- South China University of Technology
- Guangzhou
- China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| |
Collapse
|
95
|
Du XJ, Wang ZY, Wang YC. Redox-sensitive dendrimersomes assembled from amphiphilic Janus dendrimers for siRNA delivery. Biomater Sci 2018; 6:2122-2129. [DOI: 10.1039/c8bm00491a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A cationic redox-sensitive Janus dendrimer (ssJD) that self-assembles into redox-sensitive dendrimersomes (RSDs) to complex with siRNA can readily deliver siRNA into tumor cells, and then rapidly release siRNA in a reductive environment to down-regulate a targeted gene.
Collapse
Affiliation(s)
- Xiao-Jiao Du
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- P.R. China
| | - Ze-Yu Wang
- International Department
- The Affiliated High School of South China Normal University
- Guangzhou
- P.R. China
| | - Yu-Cai Wang
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- P.R. China
| |
Collapse
|
96
|
Liu Y, Zhang X, Sun T, Jiang J, Li Y, Chen M, Wei Z, Jiang W, Zhou L. Knockdown of Golgi phosphoprotein 2 inhibits hepatocellular carcinoma cell proliferation and motility. Oncotarget 2017; 7:21404-15. [PMID: 26870893 PMCID: PMC5008294 DOI: 10.18632/oncotarget.7271] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/24/2016] [Indexed: 12/31/2022] Open
Abstract
Golgi phosphoprotein 2 (GP73) is highly expressed in hepatocellular carcinoma (HCC) cells, where it serves as a biomarker and indicator of disease progression. We used MTS assays, anchorage-independent cell colony formation assays and a xenograft tumor model to show that GP73-specific siRNAs inhibit HCC proliferation in HepG2, SMMC-7721, and Huh7 cell lines and in vivo. Following GP73 silencing, levels of p-Rb, a factor related to metastasis, were reduced, but cell cycle progression was unaffected. Our results suggest that GP73 silencing may not directly suppress proliferation, but may instead inhibit cell motility. Results from proliferation assays suggest GP73 reduces expression of epithelial mesenchymal transition (EMT)-related factors and promotes cell motility, while transwell migration and invasion assays indicated a possible role in metastasis. Immunofluorescence co-localization microscopy and immunoblotting showed that GP73 decreases expression of N-cadherin and E-cadherin, two key factors in EMT, which may in turn decrease intracellular adhesive forces and promote cell motility. This study confirmed that GP73 expression leads to increased expression of EMT-related proteins and that GP73 silencing reduces HCC cell migration in vitro. These findings suggest that GP73 silencing through siRNA delivery may provide a novel low-toxicity therapy for the inhibition of tumor proliferation and metastasis.
Collapse
Affiliation(s)
- Yiming Liu
- Medical Biotechnology Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaodi Zhang
- Medical Biotechnology Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ting Sun
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Junchang Jiang
- Department of Pathology, RunRun-Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Ying Li
- Clinical Laboratory, Children's Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Mingliang Chen
- Medical Biotechnology Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhen Wei
- Laboratory Animal Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weiqin Jiang
- Cancer Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Linfu Zhou
- Medical Biotechnology Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
97
|
Sokolova V, Shi Z, Huang S, Du Y, Kopp M, Frede A, Knuschke T, Buer J, Yang D, Wu J, Westendorf AM, Epple M. Delivery of the TLR ligand poly(I:C) to liver cells in vitro and in vivo by calcium phosphate nanoparticles leads to a pronounced immunostimulation. Acta Biomater 2017; 64:401-410. [PMID: 28963016 DOI: 10.1016/j.actbio.2017.09.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 01/01/2023]
Abstract
The selective activation of the immune system is a concurrent problem in the treatment of persistent diseases like viral infections (e.g. hepatitis). For the delivery of the toll-like receptor ligand poly(I:C), an immunostimulatory action was discovered earlier by hydrodynamic injection. However, this technique is not clinically transferable to human patients. A modular system where the immunoactive toll-like-receptor ligand 3 (TLR-3) poly(I:C) was incorporated into calcium phosphate nanoparticles was developed. The nanoparticles had a hydrodynamic diameter of 275nm and a zeta potential of +20mV, measured by dynamic light scattering. The diameter of the solid core was 120nm by scanning electron microscopy. In vitro, the nanoparticle uptake was investigated after 1 and 24h of incubation of THP-1 cells (macrophages) with nanoparticles by fluorescence microscopy. After intravenous injection into BALB/c and C57BL/6J mice, respectively, the in vivo uptake was especially prominent in lung and liver, 1 and 3h after the injection. Pronounced immunostimulatory effects of the nanoparticles were found in vitro with primary liver cells, i.e. Kupffer cells (KC) and liver sinusoidal endothelial cells (LSEC) from wild-type C57BL/6J mice. Thus, they represent a suitable alternative to hydrodynamic injection treatments for future vaccination concepts. STATEMENT OF SIGNIFICANCE The selective activation of the immune system is a concurrent problem in the treatment of persistent diseases like viral infections (e.g. hepatitis). For the delivery of the toll-like receptor ligand poly(I:C), an immunostimulatory action has been discovered earlier by hydrodynamic injection. However, this technique is not clinically transferable to human patients. We have developed a modular system where poly(I:C) was incorporated into calcium phosphate nanoparticles. The uptake into relevant liver cells was studied both in vitro and in vivo. After intravenous injection into mice, the in vivo uptake was especially prominent in lung and liver, 1 and 3h after the injection. The corresponding strong immune reaction proves their high potential to turn up the immune system, e.g. against viral infections, without adverse side reactions.
Collapse
Affiliation(s)
- Viktoriya Sokolova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany
| | - Zou Shi
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Shunmei Huang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Yanqin Du
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Mathis Kopp
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany
| | - Annika Frede
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Torben Knuschke
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Jan Buer
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Jun Wu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Astrid Maria Westendorf
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany.
| |
Collapse
|
98
|
Hu J, Sheng Y, Kwak KJ, Shi J, Yu B, Lee LJ. A signal-amplifiable biochip quantifies extracellular vesicle-associated RNAs for early cancer detection. Nat Commun 2017; 8:1683. [PMID: 29162835 PMCID: PMC5698315 DOI: 10.1038/s41467-017-01942-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/26/2017] [Indexed: 11/09/2022] Open
Abstract
Detection of extracellular vesicle (EV)-associated RNAs with low expression levels in early-stage cancer remains a challenge and is highly valuable. Here, we report a nanoparticle-based biochip that could capture circulating EVs without isolation, brighten encapsulated RNAs, and amplify fluorescence signals in situ in a single step. We confine catalyzed hairpin DNA circuit (CHDC) in cationic lipid-polymer hybrid nanoparticles (LPHNs) that are tethered on a chip. LPHN features a core-shell-corona structure that facilitates the transfer and mixing of CHDC with EV-associated RNAs when forming the LPHN–EV nanocomplex. CHDC is triggered upon target RNA binding and quickly generate amplified signals. The signal amplification efficiency of LPHN–CHDC is demonstrated in artificial EVs, cancer cells, and cancer cell-derived EVs. We show that LPHN–CHDC biochip with signal amplification capability could selectively and sensitively identify low expression glypican-1 mRNA in serum EVs, distinguishing patients with early- and late-stage pancreatic cancer from healthy donors and patients with benign pancreatic disease. Extracellular vesicles (EV)-associated RNAs are serum biomarkers potentially exploitable for early cancer diagnosis. Here the authors develop a catalyzed hairpin DNA circuit within a cationic lipid-polymer hybrid nanoparticle that can detect low–level EV-associated RNAs in early stage cancer patients.
Collapse
Affiliation(s)
- Jiaming Hu
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA.
| | - Yan Sheng
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA. .,College of Chemistry and Chemical Engineering, Yantai University, Yantai, 264005, People's Republic of China.
| | - Kwang Joo Kwak
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Junfeng Shi
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Bohao Yu
- School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, People's Republic of China
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
99
|
Zou Y, Zheng M, Yang W, Meng F, Miyata K, Kim HJ, Kataoka K, Zhong Z. Virus-Mimicking Chimaeric Polymersomes Boost Targeted Cancer siRNA Therapy In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29. [PMID: 28961339 DOI: 10.1002/adma.201703285] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/02/2017] [Indexed: 05/16/2023]
Abstract
Small interfering RNA (siRNA) offers a highly selective and effective pharmaceutical for various life-threatening diseases, including cancers. The clinical translation of siRNA is, however, challenged by its short plasma life, poor cell uptake, and cumbersome intracellular trafficking. Here, cNGQGEQc peptide-functionalized reversibly crosslinked chimaeric polymersomes (cNGQ/RCCPs) is shown to mediate high-efficiency targeted delivery of Polo-like kinase1 specific siRNA (siPLK1) to orthotopic human lung cancer in nude mice. Strikingly, siRNA is completely and tightly loaded into the aqueous lumen of the polymersomes at an unprecedentedly low N/P ratio of 0.45. cNGQ/RCCPs loaded with firefly luciferase specific siRNA (siGL3) or siPLK1 are efficiently taken up by α3 β1 -integrin-overexpressing A549 lung cancer cells and quickly release the payloads to the cytoplasm, inducing highly potent and sequence-specific gene silencing in vitro. The in vivo studies using nude mice bearing orthotopic A549 human lung tumors reveal that siPLK1-loaded cNGQ/RCCPs boost long circulation, superb tumor accumulation and selectivity, effective suppression of tumor growth, and significantly improved survival time. These virus-mimicking chimaeric polymersomes provide a robust and potent platform for targeted cancer siRNA therapy.
Collapse
Affiliation(s)
- Yan Zou
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Meng Zheng
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Weijing Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8656, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, and Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hyun Jin Kim
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, and Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kazunori Kataoka
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8656, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, and Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
- Policy Alternative Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Innovation Center of NanoMedicine, Institute of Industry Promotion-Kawasaki, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
100
|
He Y, Liu P, Shi C, Liu Y, Liu S, Feng X, Fu D. The influence of hydrophilic mPEG segment on formation, morphology, and properties of PCL-mPEG microspheres. ADVANCES IN POLYMER TECHNOLOGY 2017. [DOI: 10.1002/adv.21887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yu He
- Department of Orthopaedics; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology (HUST); Wuhan China
| | - Ping Liu
- Department of Orthopaedics; Li Yuan Hospital; Tongji Medical College; Huazhong University of Science and Technology (HUST); Wuhan China
| | - Chen Shi
- Department of Pharmacy; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology (HUST); Wuhan China
| | - Yongwei Liu
- Department of Orthopaedics; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology (HUST); Wuhan China
| | - Songxiang Liu
- Department of Orthopaedics; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology (HUST); Wuhan China
| | - Xiaobo Feng
- Department of Orthopaedics; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology (HUST); Wuhan China
| | - Dehao Fu
- Department of Orthopaedics; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology (HUST); Wuhan China
| |
Collapse
|