1
|
Zou Y, Zhang J, Chen L, Xu Q, Yao S, Chen H. Targeting Neuroinflammation in Central Nervous System Diseases by Oral Delivery of Lipid Nanoparticles. Pharmaceutics 2025; 17:388. [PMID: 40143051 PMCID: PMC11944764 DOI: 10.3390/pharmaceutics17030388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Neuroinflammation within the central nervous system (CNS) is a primary characteristic of CNS diseases, such as Parkinson's disease, Alzheimer's disease (AD), amyotrophic lateral sclerosis, and mental disorders. The excessive activation of immune cells results in the massive release of pro-inflammatory cytokines, which subsequently induce neuronal death and accelerate the progression of neurodegeneration. Therefore, mitigating excessive neuroinflammation has emerged as a promising strategy for the treatment of CNS diseases. Despite advancements in drug discovery and the development of novel therapeutics, the effective delivery of these agents to the CNS remains a serious challenge due to the restrictive nature of the blood-brain barrier (BBB). This underscores the need to develop a novel drug delivery system. Recent studies have identified oral lipid nanoparticles (LNPs) as a promising approach to efficiently deliver drugs across the BBB and treat neurological diseases. This review aims to comprehensively summarize the recent advancements in the development of LNPs designed for the controlled delivery and therapeutic modulation of CNS diseases through oral administration. Furthermore, this review addresses the mechanisms by which these LNPs overcome biological barriers and evaluate their clinical implications and therapeutic efficacy in the context of oral drug delivery systems. Specifically, it focuses on LNP formulations that facilitate oral administration, exploring their potential to enhance bioavailability, improve targeting precision, and alleviate or manage the symptoms associated with a range of CNS diseases.
Collapse
Affiliation(s)
- Yuan Zou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jing Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430074, China; (J.Z.); (Q.X.)
| | - Longmin Chen
- Department of Rheumatology and Immunology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Qianqian Xu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430074, China; (J.Z.); (Q.X.)
| | - Sheng Yao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
2
|
Dorsey PJ, Lau CL, Chang TC, Doerschuk PC, D'Addio SM. Review of machine learning for lipid nanoparticle formulation and process development. J Pharm Sci 2024; 113:3413-3433. [PMID: 39341497 DOI: 10.1016/j.xphs.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024]
Abstract
Lipid nanoparticles (LNPs) are a subset of pharmaceutical nanoparticulate formulations designed to encapsulate, stabilize, and deliver nucleic acid cargoes in vivo. Applications for LNPs include new interventions for genetic disorders, novel classes of vaccines, and alternate modes of intracellular delivery for therapeutic proteins. In the pharmaceutical industry, establishing a robust formulation and process to achieve target product performance is a critical component of drug development. Fundamental understanding of the processes for making LNPs and their interactions with biological systems have advanced considerably in the wake of the COVID-19 pandemic. Nevertheless, LNP formulation research remains largely empirical and resource intensive due to the multitude of input parameters and the complex physical phenomena that govern the processes of nanoparticle precipitation, self-assembly, structure evolution, and stability. Increasingly, artificial intelligence and machine learning (AI/ML) are being applied to improve the efficiency of research activities through in silico models and predictions, and to drive deeper fundamental understanding of experimental inputs to functional outputs. This review will identify current challenges and opportunities in the development of robust LNP formulations of nucleic acids, review studies that apply machine learning methods to experimental datasets, and provide discussion on associated data science challenges to facilitate collaboration between formulation and data scientists, aiming to accelerate the advancement of AI/ML applied to LNP formulation and process optimization.
Collapse
Affiliation(s)
- Phillip J Dorsey
- Pharmaceutical Sciences & Clinical Supply, MRL, Merck & Co., Inc., Rahway, NJ 07065, USA; University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Christina L Lau
- Cornell University, School of Electrical and Computer Engineering, Ithaca, NY 14853, USA
| | - Ti-Chiun Chang
- Pharmaceutical Sciences & Clinical Supply, MRL, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Peter C Doerschuk
- Cornell University, School of Electrical and Computer Engineering, Ithaca, NY 14853, USA
| | - Suzanne M D'Addio
- Pharmaceutical Sciences & Clinical Supply, MRL, Merck & Co., Inc., Rahway, NJ 07065, USA.
| |
Collapse
|
3
|
Zhang YQ, Guo RR, Chen YH, Li TC, Du WZ, Xiang RW, Guan JB, Li YP, Huang YY, Yu ZQ, Cai Y, Zhang P, Ling GX. Ionizable drug delivery systems for efficient and selective gene therapy. Mil Med Res 2023; 10:9. [PMID: 36843103 PMCID: PMC9968649 DOI: 10.1186/s40779-023-00445-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 02/01/2023] [Indexed: 02/28/2023] Open
Abstract
Gene therapy has shown great potential to treat various diseases by repairing the abnormal gene function. However, a great challenge in bringing the nucleic acid formulations to the market is the safe and effective delivery to the specific tissues and cells. To be excited, the development of ionizable drug delivery systems (IDDSs) has promoted a great breakthrough as evidenced by the approval of the BNT162b2 vaccine for prevention of coronavirus disease 2019 (COVID-19) in 2021. Compared with conventional cationic gene vectors, IDDSs can decrease the toxicity of carriers to cell membranes, and increase cellular uptake and endosomal escape of nucleic acids by their unique pH-responsive structures. Despite the progress, there remain necessary requirements for designing more efficient IDDSs for precise gene therapy. Herein, we systematically classify the IDDSs and summarize the characteristics and advantages of IDDSs in order to explore the underlying design mechanisms. The delivery mechanisms and therapeutic applications of IDDSs are comprehensively reviewed for the delivery of pDNA and four kinds of RNA. In particular, organ selecting considerations and high-throughput screening are highlighted to explore efficiently multifunctional ionizable nanomaterials with superior gene delivery capacity. We anticipate providing references for researchers to rationally design more efficient and accurate targeted gene delivery systems in the future, and indicate ideas for developing next generation gene vectors.
Collapse
Affiliation(s)
- Yu-Qi Zhang
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Ran-Ran Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Yong-Hu Chen
- School of Pharmacy, Yanbian University, Yanji, 133002, Jilin, China
| | - Tian-Cheng Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Wen-Zhen Du
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Rong-Wu Xiang
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Ji-Bin Guan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yu-Peng Li
- Masonic Cancer Center and Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yuan-Yu Huang
- Advanced Research Institute of Multidisciplinary Science; School of Life Science; School of Medical Technology; Key Laboratory of Molecular Medicine and Biotherapy; Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhi-Qiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, Guangdong, China
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China.
| | - Gui-Xia Ling
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
4
|
Self-homing nanocarriers for mRNA delivery to the activated hepatic stellate cells in liver fibrosis. J Control Release 2023; 353:685-698. [PMID: 36521688 DOI: 10.1016/j.jconrel.2022.12.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/17/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Herein, we report on the development of a platform for the selective delivery of mRNA to the hard-to-transfect Activated Hepatic Stellate Cells (aHSCs), the fundamental player in the progression of liver fibrosis. Using a microfluidic device (iLiNP), we prepared a series of lipid nanoparticles (LNPs) based on a diverse library of pH-sensitive lipids. After an in-depth in vivo optimization of the LNPs, their mRNA delivery efficiency, selectivity, potency, robustness, and biosafety were confirmed. Furthermore, some mechanistic aspects of their selective delivery to aHSCs were investigated. We identified a promising lipid candidate, CL15A6, that has a high affinity to aHSCs. Tweaking the composition and physico-chemical properties of the LNPs enabled the robust and ligand-free mRNA delivery to aHSCs in vivo post intravenous administration, with a high biosafety at mRNA doses of up to 2 mg/Kg, upon either acute or chronic administrations. The mechanistic investigation suggested that CL15A6 LNPs were taken up by aHSCs via Clathrin-mediated endocytosis through the Platelet-derived growth factor receptor beta (PDGFRβ) and showed a pKa-dependent cellular uptake. The novel and scalable platform reported in this study is highly promising for clinical applications.
Collapse
|
5
|
Sun D, Lu ZR. Structure and Function of Cationic and Ionizable Lipids for Nucleic Acid Delivery. Pharm Res 2023; 40:27-46. [PMID: 36600047 PMCID: PMC9812548 DOI: 10.1007/s11095-022-03460-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023]
Abstract
Hereditary genetic diseases, cancer, and infectious diseases are affecting global health and become major health issues, but the treatment development remains challenging. Gene therapies using DNA plasmid, RNAi, miRNA, mRNA, and gene editing hold great promise. Lipid nanoparticle (LNP) delivery technology has been a revolutionary development, which has been granted for clinical applications, including mRNA vaccines against SARS-CoV-2 infections. Due to the success of LNP systems, understanding the structure, formulation, and function relationship of the lipid components in LNP systems is crucial for design more effective LNP. Here, we highlight the key considerations for developing an LNP system. The evolution of structure and function of lipids as well as their LNP formulation from the early-stage simple formulations to multi-components LNP and multifunctional ionizable lipids have been discussed. The flexibility and platform nature of LNP enable efficient intracellular delivery of a variety of therapeutic nucleic acids and provide many novel treatment options for the diseases that are previously untreatable.
Collapse
Affiliation(s)
- Da Sun
- Department of Biomedical Engineering, Case Western Reserve University, Wickenden 427, Mail Stop 7207, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Wickenden 427, Mail Stop 7207, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
6
|
Zhang X, Wang M, Feng J, Qin B, Zhang C, Zhu C, Liu W, Wang Y, Liu W, Huang L, Lu S, Wang Z. Multifunctional nanoparticles co-loaded with Adriamycin and MDR-targeting siRNAs for treatment of chemotherapy-resistant esophageal cancer. J Nanobiotechnology 2022; 20:166. [PMID: 35346194 PMCID: PMC8962182 DOI: 10.1186/s12951-022-01377-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/14/2022] [Indexed: 02/08/2023] Open
Abstract
The development of multidrug resistance (MDR) during cancer chemotherapy is a major challenge in current cancer treatment strategies. Numerous molecular mechanisms, including increased drug efflux, evasion of drug-induced apoptosis, and activation of DNA repair mechanisms, can drive chemotherapy resistance. Here we have identified the major vault protein (MVP) and the B-cell lymphoma-2 (BCL2) gene as two potential factors driving MDR in esophageal squamous cell carcinoma (ESCC). We have designed a novel and versatile self-assembling nanoparticle (NP) platform on a multifunctional carboxymethyl chitosan base to simultaneously deliver Adriamycin, and siRNAs targeting MVP and BCL2 (CEAMB NPs), thus reducing drug efflux and promoting apoptosis of esophageal cancer cells. To achieve effective delivery to tumor tissues and inhibit tumor growth in vivo, carboxymethyl chitosan was engineered to contain multiple histidines for enhanced cytosol delivery, cholesterol for improved self-assembly, and epidermal growth factor receptor (EGFR) antibodies to target cancer cells. Our results indicate that these nanoparticles are efficiently synthesized with the desired chemical composition to self-assemble into cargo-containing NPs. Furthermore, we have shown that the synthesized NPs will successfully inhibit cancer cells growth and tumor development when delivered to cultured ESCC cells or to in vivo mouse xenograft models. Our engineered NPs offer a potential novel platform in treating various types of chemotherapy-resistant tumors.
Collapse
Affiliation(s)
- Xiangyang Zhang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Min Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Junyi Feng
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Bin Qin
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Chenglin Zhang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Chengshen Zhu
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Wentao Liu
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yaohe Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Wei Liu
- Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Lei Huang
- Inflammations Immunity Research Theme, Translational and Clinical Research Institute, FMS, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Shuangshuang Lu
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhimin Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
7
|
Zhang X, Qin B, Wang M, Feng J, Zhang C, Zhu C, He S, Liu H, Wang Y, Averick SE, Vo NTN, Huang L, Liu W, Wang Z. Dual pH-Responsive and Tumor-Targeted Nanoparticle-Mediated Anti-Angiogenesis siRNA Delivery for Tumor Treatment. Int J Nanomedicine 2022; 17:953-967. [PMID: 35280336 PMCID: PMC8906879 DOI: 10.2147/ijn.s340926] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose In order to overcome the biological barriers at all levels and enhance the delivery efficiency of siRNA, we have prepared a multifunctional siRNA delivery system (CHCE/siRNA nanoparticles) through self-assembly of the carboxymethyl chitosan modified with histidine, cholesterol, and anti-EGFR antibody (CHCE). Methods The morphology of CHCE/siRNA NPs was detected by dynamic light scattering and scanning electron microscope. In vitro, we assessed the tumor-targeting, cellular uptake, and endosomal escape by flow cytometry and confocal laser scanning microscopy, confirming the CHCE/siRNA NPs functions in gene silencing and cell killing ability. In vivo, we examined the biodistribution of the CHCE/siRNA NPs by the IVIS imaging system and confirmed the therapeutic effect of NPs in the nude-mouse tumor model. Results The CHCE/siRNA NPs exhibited nanosized spherical with narrow size distribution. In vitro, the CHCE/siRNA NPs incorporated a dual capability of tumor targeting and pH response that could facilitate cellular bind, cellular uptake, and endosomal escape. The CHCE/siRNA NPs could effectively silence the vascular endothelial growth factor A (VEGFA) to cause cell apoptosis and inhibit proliferation. In vivo, the CHCE/siRNA NPs could target tumor sites to knock down VEGFA and achieve a better anti-tumor effect. Conclusion We successfully prepared a novel siRNA delivery system with the double capability of tumor targeting and pH response, which can break through the biological barriers to penetrate deep into tumors and achieve better therapeutic tumor effects, providing a new ideal delivery platform for siRNA.
Collapse
Affiliation(s)
- Xiangyang Zhang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Bin Qin
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Min Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Junyi Feng
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Chenglin Zhang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Chengshen Zhu
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Suqin He
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Hao Liu
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Yaohe Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Saadyah E Averick
- Neuroscience Institute, Allegheny Health Network, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Nga T N Vo
- School of Engineering, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Lei Huang
- Inflammations Immunity Research Theme, Translational and Clinical Research Institute, FMS, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Wentao Liu
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Zhimin Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Correspondence: Zhimin Wang; Wentao Liu, Email ;
| |
Collapse
|
8
|
Kheirolomoom A, Kare AJ, Ingham ES, Paulmurugan R, Robinson ER, Baikoghli M, Inayathullah M, Seo JW, Wang J, Fite BZ, Wu B, Tumbale SK, Raie MN, Cheng RH, Nichols L, Borowsky AD, Ferrara KW. In situ T-cell transfection by anti-CD3-conjugated lipid nanoparticles leads to T-cell activation, migration, and phenotypic shift. Biomaterials 2022; 281:121339. [PMID: 35078042 PMCID: PMC8892572 DOI: 10.1016/j.biomaterials.2021.121339] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 02/03/2023]
Abstract
Ex vivo programming of T cells can be efficacious but is complex and expensive; therefore, the development of methods to transfect T cells in situ is important. We developed and optimized anti-CD3-targeted lipid nanoparticles (aCD3-LNPs) to deliver tightly packed, reporter gene mRNA specifically to T cells. In vitro, targeted LNPs efficiently delivered mCherry mRNA to Jurkat T cells, and T-cell activation and depletion were associated with aCD3 antibody coating on the surface of LNPs. aCD3-LNPs, but not non-targeted LNPs, accumulated within the spleen following systemic injection, with mCherry and Fluc signals visible within 30 min after injection. At 24 h after aCD3-LNP injection, 2-4% of all splenic T cells and 2-7% of all circulating T cells expressed mCherry, and this was dependent on aCD3 coating density. Targeting and transfection were accompanied by systemic CD25+, OX40+, and CD69+ T-cell activation with temporary CD3e ligand loss and depletion of splenic and circulating subsets. Migration of splenic CD8a+ T cells from the white-pulp to red-pulp, and differentiation from naïve to memory and effector phenotypes, followed upon aCD3-LNP delivery. Additionally, aCD3-LNP injection stimulated the secretion of myeloid-derived chemokines and T-helper cytokines into plasma. Lastly, we administered aCD3-LNPs to tumor bearing mice and found that transfected T cells localized within tumors and tumor-draining lymph nodes following immunotherapy treatment. In summary, we show that CD3-targeted transfection is feasible, yet associated with complex immunological consequences that must be further studied for potential therapeutic applications.
Collapse
Affiliation(s)
| | - Aris J. Kare
- Stanford University, Department of Bioengineering,
Stanford, CA, USA
| | - Elizabeth S. Ingham
- University of California, Davis, Department of Biomedical
Engineering, Davis, CA 95616, USA
| | | | | | - Mo Baikoghli
- University of California, Davis, Department of Molecular
and Cellular Biology, Davis, CA, USA
| | | | - Jai W. Seo
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - James Wang
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - Brett Z. Fite
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - Bo Wu
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | | | - Marina N. Raie
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - R. Holland Cheng
- University of California, Davis, Department of Molecular
and Cellular Biology, Davis, CA, USA
| | - Lisa Nichols
- Stanford Shared FACS Facility, Stanford University,
Stanford, CA, USA
| | | | - Katherine W. Ferrara
- Stanford University, Department of Radiology, Palo Alto,
CA, USA,Corresponding author: Katherine W. Ferrara, PhD,
Professor and Division Chief, Molecular Imaging Program at Stanford, Department
of Radiology, 3165 Porter Drive, Stanford University, Palo Alto, CA 94304,
Phone: (650)723-8906,
| |
Collapse
|
9
|
Billingsley MM, Hamilton AG, Mai D, Patel SK, Swingle KL, Sheppard NC, June CH, Mitchell MJ. Orthogonal Design of Experiments for Optimization of Lipid Nanoparticles for mRNA Engineering of CAR T Cells. NANO LETTERS 2022; 22:533-542. [PMID: 34669421 PMCID: PMC9335860 DOI: 10.1021/acs.nanolett.1c02503] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Viral engineered chimeric antigen receptor (CAR) T cell therapies are potent, targeted cancer immunotherapies, but their permanent CAR expression can lead to severe adverse effects. Nonviral messenger RNA (mRNA) CAR T cells are being explored to overcome these drawbacks, but electroporation, the most common T cell transfection method, is limited by cytotoxicity. As a potentially safer nonviral delivery strategy, here, sequential libraries of ionizable lipid nanoparticle (LNP) formulations with varied excipient compositions were screened in comparison to a standard formulation for improved mRNA delivery to T cells with low cytotoxicity, revealing B10 as the top formulation with a 3-fold increase in mRNA delivery. When compared to electroporation in primary human T cells, B10 LNPs induced comparable CAR expression with reduced cytotoxicity while demonstrating potent cancer cell killing. These results demonstrate the impact of excipient optimization on LNP performance and support B10 LNPs as a potent mRNA delivery platform for T cell engineering.
Collapse
Affiliation(s)
- Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alex G Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David Mai
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Savan K Patel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Neil C Sheppard
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carl H June
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
10
|
Abstract
RNA-based therapeutics have shown great promise in treating a broad spectrum of diseases through various mechanisms including knockdown of pathological genes, expression of therapeutic proteins, and programmed gene editing. Due to the inherent instability and negative-charges of RNA molecules, RNA-based therapeutics can make the most use of delivery systems to overcome biological barriers and to release the RNA payload into the cytosol. Among different types of delivery systems, lipid-based RNA delivery systems, particularly lipid nanoparticles (LNPs), have been extensively studied due to their unique properties, such as simple chemical synthesis of lipid components, scalable manufacturing processes of LNPs, and wide packaging capability. LNPs represent the most widely used delivery systems for RNA-based therapeutics, as evidenced by the clinical approvals of three LNP-RNA formulations, patisiran, BNT162b2, and mRNA-1273. This review covers recent advances of lipids, lipid derivatives, and lipid-derived macromolecules used in RNA delivery over the past several decades. We focus mainly on their chemical structures, synthetic routes, characterization, formulation methods, and structure-activity relationships. We also briefly describe the current status of representative preclinical studies and clinical trials and highlight future opportunities and challenges.
Collapse
Affiliation(s)
- Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Changzhen Sun
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chang Wang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Katarina E Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
11
|
Kawase W, Kurotaki D, Suzuki Y, Ishihara H, Ban T, Sato GR, Ichikawa J, Yanai H, Taniguchi T, Tsukahara K, Tamura T. Irf5 siRNA-loaded biodegradable lipid nanoparticles ameliorate concanavalin A-induced liver injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:708-715. [PMID: 34589288 PMCID: PMC8463440 DOI: 10.1016/j.omtn.2021.08.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022]
Abstract
RNA interference-based gene silencing drugs are attracting attention for treating various diseases. Lipid nanoparticles (LNPs) are carriers that efficiently deliver small interfering RNA (siRNA) to the cytoplasm of target cells. Recently, we developed potent and well-tolerated biodegradable LNPs with asymmetric ionizable lipids. Here, we evaluated the effect of LNPs on immune cells in mice. After intravenous administration, LNPs were efficiently incorporated into several tissue-resident macrophages, including liver macrophages, through an apolipoprotein E (ApoE)-independent mechanism. Administration of LNP-encapsulated siRNA against Irf5, encoding the transcription factor critical for inflammatory responses, sharply reduced its expression in macrophages in vivo, and persisted for as long as 7 days. The therapeutic potential of Irf5 siRNA-loaded LNPs in inflammatory diseases was tested in a concanavalin A (Con A)-induced hepatitis model, whose pathogenic mechanisms are dependent on cytokine secretion from macrophages. We found that Con A-induced liver injury was significantly attenuated after LNP injection. Serum aspartate transaminase, alanine aminotransferase, and inflammatory cytokine levels were significantly reduced in mice injected with Irf5 siRNA-loaded LNPs compared to those injected with control siRNA-loaded LNPs. Our results suggest that administering biodegradable LNPs to deliver siRNA is a promising strategy for treating inflammatory disorders.
Collapse
Affiliation(s)
- Wataru Kawase
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan.,Laboratory of Chromatin Organization in Immune Cell Development, International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yuta Suzuki
- Tsukuba Research Laboratories, Eisai Co., Ltd, Tsukuba 300-2635, Japan
| | - Hiroshi Ishihara
- Tsukuba Research Laboratories, Eisai Co., Ltd, Tsukuba 300-2635, Japan
| | - Tatsuma Ban
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Go R Sato
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Juri Ichikawa
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Hideyuki Yanai
- Department of Inflammology, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-0041, Japan
| | - Tadatsugu Taniguchi
- Department of Inflammology, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-0041, Japan
| | - Kappei Tsukahara
- Tsukuba Research Laboratories, Eisai Co., Ltd, Tsukuba 300-2635, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan
| |
Collapse
|
12
|
Eygeris Y, Patel S, Jozic A, Sahay G. Deconvoluting Lipid Nanoparticle Structure for Messenger RNA Delivery. NANO LETTERS 2020; 20:4543-4549. [PMID: 32375002 PMCID: PMC7228479 DOI: 10.1021/acs.nanolett.0c01386] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/29/2020] [Indexed: 05/19/2023]
Abstract
Lipid nanoparticle (LNP) packaged mRNA vaccines have been deployed against infectious diseases such as COVID-19, yet their structural features remain unclear. Cholesterol, a major constituent within LNPs, contributes to their morphology that influences gene delivery. Herein, we examine the structure of LNPs containing cholesterol derivatives using electron microscopy, differential scanning calorimetry, and membrane fluidity assays. LNPs formulated with C24 alkyl derivatives of cholesterol show a polymorphic shape and various degrees of multilamellarity and lipid partitioning, likely due to phase separation. The addition of methyl and ethyl groups to the C24 alkyl tail of the cholesterol backbone induces multilamellarity (>50% increase compared to cholesterol), while the addition of a double bond induces lipid partitioning (>90% increase compared to cholesterol). LNPs with multilamellar and faceted structures, as well as a lamellar lipid phase, showed higher gene transfection. Unraveling the structure of mRNA-LNPs can enable their rational design toward enhanced gene delivery.
Collapse
Affiliation(s)
- Yulia Eygeris
- Department of Pharmaceutical Sciences, College of
Pharmacy, Oregon State University, Portland, Oregon 97201,
United States
| | - Siddharth Patel
- Department of Pharmaceutical Sciences, College of
Pharmacy, Oregon State University, Portland, Oregon 97201,
United States
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of
Pharmacy, Oregon State University, Portland, Oregon 97201,
United States
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of
Pharmacy, Oregon State University, Portland, Oregon 97201,
United States
- Department of Biomedical Engineering,
Oregon Health & Science University, Portland, Oregon
97201, United States
| |
Collapse
|
13
|
Eygeris Y, Patel S, Jozic A, Sahay G. Deconvoluting Lipid Nanoparticle Structure for Messenger RNA Delivery. NANO LETTERS 2020. [PMID: 32375002 DOI: 10.1021/acs.nanolett.0c0138610.1021/acs.nanolett.0c01386.s001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Lipid nanoparticle (LNP) packaged mRNA vaccines have been deployed against infectious diseases such as COVID-19, yet their structural features remain unclear. Cholesterol, a major constituent within LNPs, contributes to their morphology that influences gene delivery. Herein, we examine the structure of LNPs containing cholesterol derivatives using electron microscopy, differential scanning calorimetry, and membrane fluidity assays. LNPs formulated with C24 alkyl derivatives of cholesterol show a polymorphic shape and various degrees of multilamellarity and lipid partitioning, likely due to phase separation. The addition of methyl and ethyl groups to the C24 alkyl tail of the cholesterol backbone induces multilamellarity (>50% increase compared to cholesterol), while the addition of a double bond induces lipid partitioning (>90% increase compared to cholesterol). LNPs with multilamellar and faceted structures, as well as a lamellar lipid phase, showed higher gene transfection. Unraveling the structure of mRNA-LNPs can enable their rational design toward enhanced gene delivery.
Collapse
Affiliation(s)
- Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
| | - Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97201, United States
| |
Collapse
|
14
|
Billingsley MM, Singh N, Ravikumar P, Zhang R, June CH, Mitchell MJ. Ionizable Lipid Nanoparticle-Mediated mRNA Delivery for Human CAR T Cell Engineering. NANO LETTERS 2020; 20:1578-1589. [PMID: 31951421 PMCID: PMC7313236 DOI: 10.1021/acs.nanolett.9b04246] [Citation(s) in RCA: 361] [Impact Index Per Article: 72.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy relies on the ex vivo manipulation of patient T cells to create potent, cancer-targeting therapies, shown to be capable of inducing remission in patients with acute lymphoblastic leukemia and large B cell lymphoma. However, current CAR T cell engineering methods use viral delivery vectors, which induce permanent CAR expression and could lead to severe adverse effects. Messenger RNA (mRNA) has been explored as a promising strategy for inducing transient CAR expression in T cells to mitigate the adverse effects associated with viral vectors, but it most commonly requires electroporation for T cell mRNA delivery, which can be cytotoxic. Here, ionizable lipid nanoparticles (LNPs) were designed for ex vivo mRNA delivery to human T cells. A library of 24 ionizable lipids was synthesized, formulated into LNPs, and screened for luciferase mRNA delivery to Jurkat cells, revealing seven formulations capable of enhanced mRNA delivery over lipofectamine. The top-performing LNP formulation, C14-4, was selected for CAR mRNA delivery to primary human T cells. This platform induced CAR expression at levels equivalent to electroporation, with substantially reduced cytotoxicity. CAR T cells engineered via C14-4 LNP treatment were then compared to electroporated CAR T cells in a coculture assay with Nalm-6 acute lymphoblastic leukemia cells, and both CAR T cell engineering methods elicited potent cancer-killing activity. These results demonstrate the ability of LNPs to deliver mRNA to primary human T cells to induce functional protein expression, and indicate the potential of LNPs to enhance mRNA-based CAR T cell engineering methods.
Collapse
Affiliation(s)
- Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Nathan Singh
- Division of Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Division of Oncology, Section of Stem Cell Biology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Pranali Ravikumar
- Division of Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Rui Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carl H June
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
15
|
Moss KH, Popova P, Hadrup SR, Astakhova K, Taskova M. Lipid Nanoparticles for Delivery of Therapeutic RNA Oligonucleotides. Mol Pharm 2019; 16:2265-2277. [PMID: 31063396 DOI: 10.1021/acs.molpharmaceut.8b01290] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gene therapy is an exciting field that has the potential to address emerging scientific and therapeutic tasks. RNA-based gene therapy has made remarkable progress in recent decades. Nevertheless, efficient targeted delivery of RNA therapeutics is still a prerequisite for entering the clinics. In this review, we introduce current delivery methods for RNA gene therapeutics based on lipid nanoparticles (LNPs). We focus on the clinical appeal of recent RNA NPs and discuss existing challenges of fabrication and screening LNP candidates for effective translation into drugs of human metabolic diseases and cancer.
Collapse
Affiliation(s)
- Keith Henry Moss
- DTU Health Technology , 202 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Petya Popova
- DTU Chemistry , 206-207 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Sine R Hadrup
- DTU Health Technology , 202 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Kira Astakhova
- DTU Chemistry , 206-207 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Maria Taskova
- DTU Chemistry , 206-207 Kemitorvet , 2800 Kongens Lyngby , Denmark
| |
Collapse
|
16
|
Kuboyama T, Yagi K, Naoi T, Era T, Yagi N, Nakasato Y, Yabuuchi H, Takahashi S, Shinohara F, Iwai H, Koubara-Yamada A, Hasegawa K, Miwa A. Simplifying the Chemical Structure of Cationic Lipids for siRNA-Lipid Nanoparticles. ACS Med Chem Lett 2019; 10:749-753. [PMID: 31097994 DOI: 10.1021/acsmedchemlett.8b00652] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/05/2019] [Indexed: 01/12/2023] Open
Abstract
We report a potent cationic lipid, SST-02 ((3-hydroxylpropyl)dilinoleylamine), which possesses a simple chemical structure and is synthesized just in one step. Cationic lipids are key components of siRNA-lipid nanoparticles (LNP), which may serve as potential therapeutic agents for various diseases. For a decade, chemists have given enhanced potency and new functions to cationic lipids along with structural complexity. In this study, we conducted a medicinal chemistry campaign pursuing chemical simplicity and found that even dilinoleylmethylamine (SST-01) and methylpalmitoleylamine could be used for the in vitro and in vivo siRNA delivery. Further optimization revealed that a hydroxyl group boosted potency, and SST-02 showed an ID50 of 0.02 mg/kg in the factor VII (FVII) model. Rats administered with 3 mg/kg of SST-02 LNP did not show changes in body weight, blood chemistry, or hematological parameters, while the AST level decreased at a dose of 5 mg/kg. The use of SST-02 avoids a lengthy synthetic route and may thus decrease the future cost of nucleic acid therapeutics.
Collapse
Affiliation(s)
- Takeshi Kuboyama
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Kaori Yagi
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Tomoyuki Naoi
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Tomohiro Era
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Nobuhiro Yagi
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Yoshisuke Nakasato
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Hayato Yabuuchi
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Saori Takahashi
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Fumikazu Shinohara
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Hiroto Iwai
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Ayumi Koubara-Yamada
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Kazumasa Hasegawa
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Atsushi Miwa
- Research Functions Unit, R&D Division, Kyowa Hakko Kirin, Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| |
Collapse
|
17
|
Understanding structure-activity relationships of pH-sensitive cationic lipids facilitates the rational identification of promising lipid nanoparticles for delivering siRNAs in vivo. J Control Release 2019; 295:140-152. [PMID: 30610950 DOI: 10.1016/j.jconrel.2019.01.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/21/2018] [Accepted: 01/01/2019] [Indexed: 12/21/2022]
Abstract
Lipid nanoparticles (LNPs) are one of the more promising technologies for efficiently delivering short interfering RNA (siRNA) in vivo. A pH-sensitive cationic lipid that facilitates the targeting of hepatocytes and endosomal escape, strongly influences the availability of siRNA, thus making it a key material for efficient siRNA delivery. A systematic knowledge regarding lipid structure-activity relationships would greatly facilitate the development of sophisticated pH-sensitive cationic lipids for use in siRNA-based therapeutics. The systemic derivatization of a hydrophilic head group and hydrophobic tails of YSK12-C4, a pH-sensitive cationic lipid that was developed in our laboratory, revealed that hydrophilic head significantly affected the apparent pKa of the final product, a key factor in both intrahepatic distribution and endosomal escape. The clogP value of a hydrophilic head group was found to be associated with the apparent pKa of the product. In contrast, the hydrophobic tail structure strongly affected intrahepatic distribution without depending on apparent pKa. A structure-activity relationship study enabled the selection of an adequate combination of a hydrophilic head group and hydrophobic tails and permitted a potent LNP composed of a pH-sensitive cationic lipid CL4H6 (CL4H6-LNPs) to be developed that showed efficient gene silencing activity (50% effective dose: 0.0025 mg/kg), biodegradability and was tolerated. In vivo experiments revealed that the CL4H6-LNPs showed a superior efficiency for endosomal escape, cytosolic release and the RNA-induced silencing for the complex-loading of siRNAs compared to the previously developed LNPs.
Collapse
|
18
|
Viger-Gravel J, Schantz A, Pinon AC, Rossini AJ, Schantz S, Emsley L. Structure of Lipid Nanoparticles Containing siRNA or mRNA by Dynamic Nuclear Polarization-Enhanced NMR Spectroscopy. J Phys Chem B 2018; 122:2073-2081. [DOI: 10.1021/acs.jpcb.7b10795] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jasmine Viger-Gravel
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | | | - Arthur C. Pinon
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Aaron J. Rossini
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
- Department
of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| | | | - Lyndon Emsley
- Institut
des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
19
|
Sun XK, Zhou J, Zhang L, Ma T, Wang YH, Yang YM, Tang YT, Li H, Wang LJ. Down-regulation of Noggin and miR-138 coordinately promote osteogenesis of mesenchymal stem cells. J Mol Histol 2017; 48:427-436. [PMID: 29094227 DOI: 10.1007/s10735-017-9740-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/22/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) can differentiate to osteocytes under suitable conditions. In recent years, micro-nucleotides have been progressively used to modulate gene expression in cells due to the consideration of safety. Our present study aimed to investigate whether co-delivery of Noggin-siRNA and antimiR-138 enhances the osteogenic effect of MSCs. Using a murine MSC line, C3H/10T1/2 cells, the delivery efficiency of Noggin-siRNA and antimiR-138 into MSCs was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). Cell phenotype and proliferation capacity was assessed by flow cytometry and MTT assay respectively. The osteogenesis of MSCs was tested by Alkaline Phosphatase (ALP) staining, qRT-PCR, and western blot analyses. Our results demonstrated that the expression of Noggin and miR-138 were significantly silenced in MSCs by Noggin-siRNA and/or antimiR-138 delivery, while the phenotype and proliferation capacity of MSCs were not affected. Down-regulation of Noggin and miR-138 cooperatively promoted osteogenic differentiation of MSCs. The ALP positive cells reached about 83.57 ± 10.18%. Compared with single delivery, the expression of osteogenic related genes, such as Alp, Col-1, Bmp2, Ocn and Runx2, were the highest in cells with co-delivery of the two oligonucleotides. Moreover, the protein level of RUNX2, and the ratios of pSMAD1/5/SMAD1/5 and pERK1/2/ERK1/2 were significantly increased. The activation of Smad, Erk signaling may constitute the underlying mechanism of the enhanced osteogenesis process. Taken together, our study provides a safe strategy for the clinical rehabilitation application of MSCs in skeletal deficiency.
Collapse
Affiliation(s)
- Xing-Kun Sun
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, 100850, China
- Department of Stomatology, General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, China
- Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
| | - Jin Zhou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, 100850, China
| | - Lei Zhang
- School of Biological and Chemical Engineering, ZheJiang University of Science & Technology, Hangzhou, 310023, China
| | - Tian Ma
- Department of Plastic and Reconstructive Surgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu-Han Wang
- Tibet Vocational Technical College, Lhasa, 850032, Tibet Autonomous Region, China
| | - Yan-Mei Yang
- Department of Stomatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan-Ting Tang
- Department of Stomatology, People's Hospital of Suzhou High-tech Zone, Suzhou, 215129, Jiangsu Province, China
| | - Hong Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, 100850, China.
| | - Li-Jun Wang
- Department of Stomatology, General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, China.
| |
Collapse
|
20
|
Abstract
RNA is emerging as a potential therapeutic modality for the treatment of incurable diseases. Despite intense research, the advent to clinical utility remains compromised by numerous biological barriers, hence, there is a need for sophisticated delivery vehicles. In this aspect, lipid nanoparticles (LNPs) are the most advanced platform among nonviral vectors for gene delivery. In this review, we critically review the literature and the reasons for ineffective delivery beyond the liver. We discuss the toxicity issues associated with permanently charged cationic lipids and then turn our attention to next-generation ionizable cationic lipids. These lipids exhibit reduced toxicity and immunogenicity and undergo ionization under the acidic environment of the endosome to release the encapsulated payload to their site of action in the cytosol. Finally, we summarize recent achievements in therapeutic nucleic acid delivery and report on the current status of clinical trials using LNP and the obstacles to clinical translation.
Collapse
Affiliation(s)
- Stephanie Rietwyk
- Laboratory of Precision NanoMedicine, Department of Cell Research & Immunology, George S. Wise Faculty of Life Sciences, ‡Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, §Center for Nanoscience and Nanotechnology, and ∥Cancer Biology Research Center, Tel Aviv University , Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Department of Cell Research & Immunology, George S. Wise Faculty of Life Sciences, ‡Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, §Center for Nanoscience and Nanotechnology, and ∥Cancer Biology Research Center, Tel Aviv University , Tel Aviv 69978, Israel
| |
Collapse
|
21
|
Trinh T, Chidchob P, Bazzi HS, Sleiman HF. DNA micelles as nanoreactors: efficient DNA functionalization with hydrophobic organic molecules. Chem Commun (Camb) 2016; 52:10914-7. [PMID: 27533528 DOI: 10.1039/c6cc04970b] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We report a micelle-templated method to enhance the reactivity of DNA with highly hydrophobic molecules. Lipids, chromophores and polymers can be conjugated to DNA in high yield and under mild conditions. This method expands the range of DNA-templated reactions for DNA-encoded libraries, oligonucleotide and drug delivery, nanopore mimetics and DNA nanotechnology.
Collapse
Affiliation(s)
- Tuan Trinh
- Department of Chemistry and Center for Self-Assembled Chemical Structures, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A 0B8.
| | | | | | | |
Collapse
|
22
|
Suzuki Y, Hyodo K, Tanaka Y, Ishihara H. siRNA-lipid nanoparticles with long-term storage stability facilitate potent gene-silencing in vivo. J Control Release 2015; 220:44-50. [DOI: 10.1016/j.jconrel.2015.10.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/28/2015] [Accepted: 10/14/2015] [Indexed: 12/18/2022]
|
23
|
Ercole F, Whittaker MR, Quinn JF, Davis TP. Cholesterol Modified Self-Assemblies and Their Application to Nanomedicine. Biomacromolecules 2015; 16:1886-914. [DOI: 10.1021/acs.biomac.5b00550] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Francesca Ercole
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Michael R. Whittaker
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - John F. Quinn
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Thomas P. Davis
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department
of Chemistry, University of Warwick, Coventry, ULCV4 7AL, United Kingdom
| |
Collapse
|
24
|
Zhang Y, Arrington L, Boardman D, Davis J, Xu Y, DiFelice K, Stirdivant S, Wang W, Budzik B, Bawiec J, Deng J, Beutner G, Seifried D, Stanton M, Gindy M, Leone A. The development of an in vitro assay to screen lipid based nanoparticles for siRNA delivery. J Control Release 2013; 174:7-14. [PMID: 24240015 DOI: 10.1016/j.jconrel.2013.11.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 10/13/2013] [Accepted: 11/04/2013] [Indexed: 12/12/2022]
Abstract
In order to rapidly screen and select lead candidates for in vivo evaluation of lipid nanoparticles (LNPs) for systemic small interfering RNA (siRNA) delivery, an in vitro assay amenable to high-throughput screening (HTS) is developed. The strategy is to mimic the in vivo experience of LNPs after systemic administration, such as interactions with serum components, exposure to endosomal pH environments, and interactions with endosomal membrane lipids. It is postulated that the amount of siRNA released from LNPs after going through these treatments can be used as a screening tool to rank order the effectiveness of siRNA delivery by lipid nanoparticles in vivo. LNPs were incubated with 50% serum from different species (i.e. mouse, rat, or rhesus) at 37°C. The resulting samples were then reacted with anionic, endosomal-mimicking lipids at different pHs. The amount of siRNA released from LNPs was determined using spectrophotometry employing the fluorescent indicator SYBR Gold. Our results indicated that the amount of siRNA liberated was highly dependent upon the species of serum used and the pH to which it was exposed. LNPs treated with mouse serum showed higher levels of siRNA release, as did those subjected to endosomal pH (6.0), compared to physiological pH. Most interestingly, a good correlation between the amount of siRNA released and the in vivo efficacy was observed. In conclusion, an in vitro siRNA release assay was developed to screen and rank order LNPs for in vivo evaluation.
Collapse
Affiliation(s)
- Ye Zhang
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA.
| | - Leticia Arrington
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - David Boardman
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Jared Davis
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Yan Xu
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Katie DiFelice
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Steve Stirdivant
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Weimin Wang
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Brian Budzik
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Jack Bawiec
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - James Deng
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Greg Beutner
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Darla Seifried
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Matthew Stanton
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Marian Gindy
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| | - Anthony Leone
- Department of RNAi Therapeutics, Merck Research Laboratories, 770 Sumneytown Pike, West Point, 19486, USA
| |
Collapse
|
25
|
Bogomolova A, Hruby M, Panek J, Rabyk M, Turner S, Bals S, Steinhart M, Zhigunov A, Sedlacek O, Stepanek P, Filippov SK. Small-angle X-ray scattering and light scattering study of hybrid nanoparticles composed of thermoresponsive triblock copolymer F127 and thermoresponsive statistical polyoxazolines with hydrophobic moieties. J Appl Crystallogr 2013. [DOI: 10.1107/s0021889813027064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A combination of new thermoresponsive statistical polyoxazolines, poly[(2-butyl-2-oxazoline)-stat-(2-isopropyl-2-oxazoline)] [pBuOx-co-piPrOx], with different hydrophobic moieties and F127 surfactant as a template system for the creation of thermosensitive nanoparticles for radionuclide delivery has recently been tested [Pánek, Filippov, Hrubý, Rabyk, Bogomolova, Kučka & Stěpánek (2012).Macromol. Rapid Commun.33, 1683–1689]. It was shown that the presence of the thermosensitive F127 triblock copolymer in solution reduces nanoparticle size and polydispersity. This article focuses on a determination of the internal structure and solution properties of the nanoparticles in the temperature range from 288 to 312 K. Here, it is demonstrated that below the cloud point temperature (CPT) the polyoxazolines and F127 form complexes that co-exist in solution with single F127 molecules and large aggregates. When the temperature is raised above the CPT, nanoparticles composed of polyoxazolines and F127 are predominant in solution. These nanoparticles could be described by a spherical shell model. It was found that the molar weight and hydrophobicity of the polymer do not influence the size of the outer radius and only slightly change the inner radius of the nanoparticles. At the same time, molar weight and hydrophobicity did affect the process of nanoparticle formation. In conclusion, poly(2-oxazoline) molecules are fully incorporated inside of F127 micelles, and this result is very promising for the successful application of such systems in radionuclide delivery.
Collapse
|
26
|
Sheng R, Luo T, Li H, Sun J, Wang Z, Cao A. 'Click' synthesized sterol-based cationic lipids as gene carriers, and the effect of skeletons and headgroups on gene delivery. Bioorg Med Chem 2013; 21:6366-77. [PMID: 24063908 DOI: 10.1016/j.bmc.2013.08.047] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/15/2013] [Accepted: 08/24/2013] [Indexed: 01/01/2023]
Abstract
In this work, we have successfully prepared a series of new sterol-based cationic lipids (1-4) via an efficient 'Click' chemistry approach. The pDNA binding affinity of these lipids was examined by EB displacement and agarose-gel retardant assay. The average particle sizes and surface charges of the sterol-based cationic lipids/pDNA lipoplexes were analyzed by dynamic laser light scattering instrument (DLS), and the morphologies of the lipoplexes were observed by atomic force microscopy (AFM). The cytotoxicity of the lipids were examined by MTT and LDH assay, and the gene transfection efficiencies of these lipid carriers were investigated by luciferase gene transfection assay in various cell lines. In addition, the intracellular uptake and trafficking/localization behavior of the Cy3-DNA loaded lipoplexes were preliminarily studied by fluorescence microscopy. The results demonstrated that the pDNA loading capacity, lipoplex particle size, zeta potential and morphology of the sterol lipids/pDNA lipoplexes depended largely on the molecular structure factors including sterol-skeletons and headgroups. Furthermore, the sterol-based lipids showed quite different cytotoxicity and gene transfection efficacy in A549 and HeLa cells. Interestingly, it was found that the cholesterol-bearing lipids 1 and 2 showed 7-10(4) times higher transfection capability than their lithocholate-bearing counterparts 3 and 4 in A549 and HeLa cell lines, suggested that the gene transfection capacity strongly relied on the structure of sterol skeletons. Moreover, the study on the structure-activity relationships of these sterol-based cationic lipid gene carriers provided a possible approach for developing low cytotoxic and high efficient lipid gene carriers by selecting suitable sterol hydrophobes and cationic headgroups.
Collapse
Affiliation(s)
- Ruilong Sheng
- Laboratory for Polymer Materials, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
27
|
The distribution and cell uptake of ApoA1 modified lipid carriers of siRNA in mouse liver in vivo. Asian J Pharm Sci 2013. [DOI: 10.1016/j.ajps.2013.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
28
|
Multiparametric approach for the evaluation of lipid nanoparticles for siRNA delivery. Proc Natl Acad Sci U S A 2013; 110:12881-6. [PMID: 23882076 DOI: 10.1073/pnas.1306529110] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nanoparticle-mediated siRNA delivery is a complex process that requires transport across numerous extracellular and intracellular barriers. As such, the development of nanoparticles for efficient delivery would benefit from an understanding of how parameters associated with these barriers relate to the physicochemical properties of nanoparticles. Here, we use a multiparametric approach for the evaluation of lipid nanoparticles (LNPs) to identify relationships between structure, biological function, and biological activity. Our results indicate that evaluation of multiple parameters associated with barriers to delivery such as siRNA entrapment, pKa, LNP stability, and cell uptake as a collective may serve as a useful prescreening tool for the advancement of LNPs in vivo. This multiparametric approach complements the use of in vitro efficacy results alone for prescreening and improves in vitro-in vivo translation by minimizing false negatives. For the LNPs used in this work, the evaluation of multiple parameters enabled the identification of LNP pKa as one of the key determinants of LNP function and activity both in vitro and in vivo. It is anticipated that this type of analysis can aid in the identification of meaningful structure-function-activity relationships, improve the in vitro screening process of nanoparticles before in vivo use, and facilitate the future design of potent nanocarriers.
Collapse
|
29
|
Gooding M, Browne LP, Quinteiro FM, Selwood DL. siRNA delivery: from lipids to cell-penetrating peptides and their mimics. Chem Biol Drug Des 2013; 80:787-809. [PMID: 22974319 DOI: 10.1111/cbdd.12052] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To deliver siRNA for therapeutic use, several hurdles must be addressed. Metabolic degradation must be blocked, and the RNAi cellular machinery is located in the cytoplasm, while double-stranded siRNA is large, highly charged and impermeable to cell membranes. To date, the solutions to the delivery issues have mostly involved different forms of lipid particle encapsulation. Cell-penetrating peptides and their mimics or analogues offer a different approach and this is an emerging field with the first in vivo examples now reported. Recent reports point to lipid receptors being involved in the cellular uptake of both types of transporter. This review examines the delivery of siRNA with a focus on cell-penetrating peptides and their small molecule and oligomeric mimics. The current status of siRNA delivery methods in clinical trials is examined. It now seems that the goal of delivering siRNA therapeutically is achievable but will they form part of a sustainable healthcare portfolio for the future.
Collapse
Affiliation(s)
- Matt Gooding
- The Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | | | | | | |
Collapse
|
30
|
Multilineage differentiation of human-derived dermal fibroblasts transfected with genes coated on PLGA nanoparticles plus growth factors. Biomaterials 2013; 34:582-97. [DOI: 10.1016/j.biomaterials.2012.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 10/01/2012] [Indexed: 01/04/2023]
|
31
|
Zhang J, Pei Y, Zhang H, Wang L, Arrington L, Zhang Y, Glass A, Leone AM. Assessing the heterogeneity level in lipid nanoparticles for siRNA delivery: size-based separation, compositional heterogeneity, and impact on bioperformance. Mol Pharm 2012; 10:397-405. [PMID: 23210488 DOI: 10.1021/mp3005337] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A primary consideration when developing lipid nanoparticle (LNP) based small interfering RNA (siRNA) therapeutics is formulation polydispersity or heterogeneity. The level of heterogeneity of physicochemical properties within a pharmaceutical batch could greatly affect the bioperformance, quality, and ability of a manufacturer to consistently control and reproduce the formulations. This article studied the heterogeneity in the size, composition, and in vitro performance of siRNA containing LNPs, by conducting preparative scale fractionation using a sephacryl S-1000 based size-exclusion chromatography (SEC) method. Eight LNPs with size in the range of 60-190 nm were first evaluated by the SEC method for size polydispersity characterization, and it was found that LNPs in the range of 60-150 nm could be well-resolved. Two LNPs (LNP A and LNP B) with similar bulk properties were fractionated, and fractions were studied in-depth for potential presence of polydispersity in size, composition, and in vitro silencing, as well as cytotoxicity. LNP A was deemed to be monodisperse following results of a semipreparative SEC fractionation that showed similar size, chemical composition, in vitro silencing activity, and cytotoxicity across the fractions. Therefore, LNP A represents a relatively homogeneous formulation and offers less of a challenge in its pharmaceutical development. In contrast, LNP B fractions were shown to be significantly more polydisperse in size distribution. Interestingly, LNP B SEC fractions also exhibited profound compositional variations (e.g., 5 fold difference in N/P ratio and 3 fold difference in lipid composition) along with up to 40 fold differences in the in vitro silencing activity. The impact of LNP size and formulation composition on in vitro performance is also discussed. The present results demonstrate the complexity and potential for presence of heterogeneity in LNP-based siRNA drug products. This underscores the need for tools that yield a detailed characterization of LNP formulations. This capability in tandem with the pursuit of improved formulation and process design can lead to more facile development of LNP-based siRNA pharmaceuticals of higher quality.
Collapse
Affiliation(s)
- Jingtao Zhang
- Department of Pharmaceutical Sciences, Merck Research Laboratories, Merck & Co, Inc, West Point, Pennsylvania 19486, United States.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Walsh CL, Nguyen J, Tiffany MR, Szoka FC. Synthesis, characterization, and evaluation of ionizable lysine-based lipids for siRNA delivery. Bioconjug Chem 2012; 24:36-43. [PMID: 23176544 DOI: 10.1021/bc300346h] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We report the synthesis and characterization of a series of ionizable lysine-based lipids (ILL), novel lipids containing a lysine headgroup linked to a long-chain dialkylamine through an amide linkage at the lysine α-amine. These ILLs contain two ionizable amines and a carboxylate, and exhibit pH-dependent lipid ionization that varies with lipid structure. The synthetic scheme employed allows for the simple, orthogonal manipulation of lipids. This provides a method for the development of a compositionally diverse library with varying ionizable headgroups, tail structures, and linker regions. A focused library of four ILLs was synthesized to determine the impact of hydrophobic fluidity, lipid net charge, and lipid pK(a) on the biophysical and siRNA transfection characteristics of this new class of lipids. We found that manipulation of lipid structure impacts the protonation behavior, electrostatically driven membrane disruption, and ability to promote siRNA mediated knockdown in vitro. ILL-siRNA liposomal formulations were tested in a murine Factor VII model; however, no significant siRNA-mediated knockdown was observed. These results indicate that ILL may be useful in vitro transfection reagents, but further optimization of this new class of lipids is required to develop an effective in vivo siRNA delivery system.
Collapse
Affiliation(s)
- Colin L Walsh
- The UC Berkeley-UCSF Graduate Program in Bioengineering, University of California Berkeley, Berkeley, CA 94720-1762, USA
| | | | | | | |
Collapse
|
33
|
Daka A, Peer D. RNAi-based nanomedicines for targeted personalized therapy. Adv Drug Deliv Rev 2012; 64:1508-21. [PMID: 22975009 DOI: 10.1016/j.addr.2012.08.014] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 08/07/2012] [Accepted: 08/13/2012] [Indexed: 12/31/2022]
Abstract
RNA interference (RNAi) has just made it through the pipeline to clinical trials. However, in order for RNAi to serve as an ideal personalized therapeutics and be clinically approved-safe, specific, and potent strategies must be devised for efficient delivery of RNAi payloads to specific cell types, which despite the immense potential, remains a challenge. Through evaluating the recent reported studies in this field, we introduce the progress in designing targeted nano-scaled strategies that are anticipated to overcome the delivery drawbacks and along with the exciting "omics" discipline to personalize RNAi-based therapeutics.
Collapse
Affiliation(s)
- Ala Daka
- Laboratory of Nanomedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Science, Israel
| | | |
Collapse
|
34
|
Zhang J, Haas RM, Leone AM. Polydispersity Characterization of Lipid Nanoparticles for siRNA Delivery Using Multiple Detection Size-Exclusion Chromatography. Anal Chem 2012; 84:6088-96. [PMID: 22816783 DOI: 10.1021/ac3007768] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Jingtao Zhang
- Department of Pharmaceutical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - R. Matthew Haas
- Department of Pharmaceutical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Anthony M. Leone
- Department of Pharmaceutical Sciences, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
35
|
Jeon SY, Park JS, Yang HN, Woo DG, Park KH. Co-delivery of SOX9 genes and anti-Cbfa-1 siRNA coated onto PLGA nanoparticles for chondrogenesis of human MSCs. Biomaterials 2012; 33:4413-23. [DOI: 10.1016/j.biomaterials.2012.02.051] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 02/27/2012] [Indexed: 01/09/2023]
|
36
|
Fujii S, Nishimura T, Sakurai K. Thermodynamics of Lipoplex Formation: Relationship between the Lipid Alkyl Tail Length and Thermodynamic Functions. CHEM LETT 2012. [DOI: 10.1246/cl.2012.501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shota Fujii
- Department of Chemistry and Biochemistry, The University of Kitakyushu
| | - Tomoki Nishimura
- Department of Chemistry and Biochemistry, The University of Kitakyushu
| | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, The University of Kitakyushu
| |
Collapse
|
37
|
Kulkarni A, DeFrees K, Hyun SH, Thompson DH. Pendant polymer:amino-β-cyclodextrin:siRNA guest:host nanoparticles as efficient vectors for gene silencing. J Am Chem Soc 2012; 134:7596-9. [PMID: 22545899 DOI: 10.1021/ja300690j] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A novel siRNA delivery vector has been developed, based on the self-assembly of monosubstituted cationic β-CD derivatives with a poly(vinyl alcohol)MW27kD (PVA) main-chain polymer bearing poly(ethylene glycol)MW2000 (PEG) and acid-labile cholesterol-modified (Chol) grafts through an acid-sensitive benzylidene acetal linkage. These components were investigated for their ability to form nanoparticles with siRNA using two different assembly schemes, involving either precomplexation of the pendant Chol-PVA-PEG polymer with the cationic β-CD derivatives before siRNA condensation or siRNA condensation with the cationic β-CD derivatives prior to addition of Chol-PVA-PEG to engage host:guest complexation. The pendant polymer:amino-β-CD:siRNA complexes were shown to form nanoparticles in the size range of 120-170 nm, with a slightly negative zeta potential. Cell viability studies in CHO-GFP cells shows that these materials have 10(3)-fold lower cytotoxicities than 25 kD bPEI, while maintaining gene-silencing efficiencies that are comparable to those of benchmark transfection reagents such as bPEI and Lipofectamine 2000. These results suggest that the degradable Chol-PVA-PEG polymer is able to self-assemble in the presence of siRNA and cationic-β-CD to form nanoparticles that are an effective and low-toxicity vehicle for delivering siRNA cargo to target cells.
Collapse
Affiliation(s)
- Aditya Kulkarni
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, USA
| | | | | | | |
Collapse
|
38
|
Aliabadi HM, Landry B, Sun C, Tang T, Uludağ H. Supramolecular assemblies in functional siRNA delivery: Where do we stand? Biomaterials 2012; 33:2546-69. [DOI: 10.1016/j.biomaterials.2011.11.079] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 11/26/2011] [Indexed: 12/14/2022]
|