1
|
Wei L, Wang H, Özkan M, Damian-Buda AI, Loynachan CN, Liao S, Stellacci F. Efficient Direct Cytosolic Protein Delivery via Protein-Linker Co-engineering. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27858-27870. [PMID: 40302608 PMCID: PMC12086766 DOI: 10.1021/acsami.5c02360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025]
Abstract
Protein therapeutics have enormous potential for transforming the treatment of intracellular cell disorders, such as genetic disorders and cancers. Due to proteins' cell-membrane impermeability, protein-based drugs against intracellular targets require efficient cytosolic delivery strategies; however, none of the current approaches are optimal. Here, we present a simple approach to render proteins membrane-permeable. We use arginine-mimicking ligand N,N'-dimethyl-1,3-propanediamine (DMPA) to functionalize the surface of a few representative proteins, varying in isoelectric point and molecular weight. We show that when these proteins have a sufficient number of these ligands on their surface, they acquire the property of penetrating the cell cytosol. Uptake experiments at 37 and 4 °C indicate that one of the penetration pathways is energy independent, with no evidence of pore formation, with inhibition assays indicating the presence of other uptake pathways. Functional tests demonstrate that the modified proteins maintain their main cellular function; specifically, modified ovalbumin (OVA) leads to enhanced antigen presentation and modified cytochrome C (Cyto C) leads to enhanced cell apoptosis. We modified bovine serum albumin (BSA) with ligands featuring different hydrophobicity and end group charges and showed that, to confer cytosolic penetration, the ligands must be cationic and that some hydrophobic content improves the penetration efficiency. This study provides a simple strategy for efficiently delivering proteins directly to the cell cytosol and offers important insights into the design and development of arginine-rich cell-penetrating peptide mimetic small molecules for protein transduction.
Collapse
Affiliation(s)
- Lixia Wei
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Heyun Wang
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Melis Özkan
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Andrada-Ioana Damian-Buda
- Institute
of Biomaterials, Department Materials Science and Engineering, Friedrich-Alexander-Universität, Erlangen 91054, Germany
| | - Colleen N. Loynachan
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Suiyang Liao
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Francesco Stellacci
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| |
Collapse
|
2
|
Liu J, Sun W, Xiao B, Xu H, Fan J, Shi X, Pan Y, Wei Q, Li R, Wang H, Piao Y, Xiang J, Shao S, Zhou Z, Shen Y, Tang J. Ionizable Lipids with Branched Linkers Enhance the Delivery of mRNA Vaccines. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13552-13564. [PMID: 39979210 DOI: 10.1021/acsami.4c21289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
The emergence of mRNA vaccines has heralded an epoch in disease prevention and treatment. Safe and efficient mRNA delivery systems are highly desired for the widespread application of mRNA therapeutics. Herein, we have designed a facile synthetic pathway for producing ionizable lipids featuring various branched linkers. These lipids have been integrated into lipid nanoparticles (LNPs) to improve the delivery of mRNA vaccines. The influence of linker structure on lipids and LNPs is currently underreported, yet it undeniably exerts a substantial impact on the outcomes. Through systematic screening and formulation optimization, we have identified that LNPs comprising ionizable lipids with a branched β-isobutylglutarate linker (bLNPs) exhibited superior transfection capabilities. In preclinical cancer prevention and treatment models, mRNA vaccines delivered by bLNPs (mRNA-bLNPs) have shown significant efficacy without causing systemic toxicity, highlighting the potential of bLNPs for clinical translation. Our synthetic strategy facilitates the expansion of the LNP library and provides valuable insights into the relationship between linker structures and delivery efficiency, thereby propelling the advancement of LNP technology.
Collapse
Affiliation(s)
- Jiwei Liu
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Wenjing Sun
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang 311200, P. R. China
| | - Bing Xiao
- Institute of Pharmaceutics, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Haoran Xu
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Jiaqi Fan
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Xueying Shi
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Yixuan Pan
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Qi Wei
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Ruoshui Li
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Huimin Wang
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang 311200, P. R. China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
- Institute of Pharmaceutics, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang 311200, P. R. China
| |
Collapse
|
3
|
Yuan Y, Li Y, Li G, Lei L, Huang X, Li M, Yao Y. Intelligent Design of Lipid Nanoparticles for Enhanced Gene Therapeutics. Mol Pharm 2025; 22:1142-1159. [PMID: 39878334 DOI: 10.1021/acs.molpharmaceut.4c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Lipid nanoparticles (LNPs) are an effective delivery system for gene therapeutics. By optimizing their formulation, the physiochemical properties of LNPs can be tailored to improve tissue penetration, cellular uptake, and precise targeting. The application of these targeted delivery strategies within the LNP framework ensures efficient delivery of therapeutic agents to specific organs or cell types, thereby maximizing therapeutic efficacy. In the realm of genome editing, LNPs have emerged as a potent vehicle for delivering CRISPR/Cas components, offering significant advantages such as high in vivo efficacy. The incorporation of machine learning into the optimization of LNP platforms for gene therapeutics represents a significant advancement, harnessing its predictive capabilities to substantially accelerate the research and development process. This review highlights the dynamic evolution of LNP technology, which is expected to drive transformative progress in the field of gene therapy.
Collapse
Affiliation(s)
- Yichen Yuan
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Ying Li
- Research Center for Space Computing System, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Guo Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
| | - Liqun Lei
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Xingxu Huang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yuan Yao
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
- Zhejiang Key Laboratory of Intelligent Manufacturing for Functional Chemicals, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| |
Collapse
|
4
|
Chen Z, Gong Y, Chen F, Lee HJ, Qian J, Zhao J, Zhang W, Li Y, Zhou Y, Xu Q, Xia Y, Zhou L, Cheng J. Orchestrated desaturation reprogramming from stearoyl-CoA desaturase to fatty acid desaturase 2 in cancer epithelial-mesenchymal transition and metastasis. Cancer Commun (Lond) 2025; 45:245-280. [PMID: 39722173 PMCID: PMC11947613 DOI: 10.1002/cac2.12644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Adaptative desaturation in fatty acid (FA) is an emerging hallmark of cancer metabolic plasticity. Desaturases such as stearoyl-CoA desaturase (SCD) and fatty acid desaturase 2 (FADS2) have been implicated in multiple cancers, and their dominant and compensatory effects have recently been highlighted. However, how tumors initiate and sustain their self-sufficient FA desaturation to maintain phenotypic transition remains elusive. This study aimed to explore the molecular orchestration of SCD and FADS2 and their specific reprogramming mechanisms in response to cancer progression. METHODS The potential interactions between SCD and FADS2 were explored by bioinformatics analyses across multiple cancer cohorts, which guided subsequent functional and mechanistic investigations. The expression levels of desaturases were investigated with online datasets and validated in both cancer tissues and cell lines. Specific desaturation activities were characterized through various isomer-resolved lipidomics methods and sensitivity assays using desaturase inhibitors. In-situ lipid profiling was conducted using multiplex stimulated Raman scattering imaging. Functional assays were performed both in vitro and in vivo, with RNA-sequencing employed for the mechanism verification. RESULTS After integration of the RNA-protein-metabolite levels, the data revealed that a reprogramming from SCD-dependent to FADS2-dependent desaturation was linked to cancer epithelial-mesenchymal transition (EMT) and progression in both patients and cell lines. FADS2 overexpression and SCD suppression concurrently maintained EMT plasticity. A FADS2/β-catenin self-reinforcing feedback loop facilitated the degree of lipid unsaturation, membrane fluidity, metastatic potential and EMT signaling. Moreover, SCD inhibition triggered a lethal apoptosis but boosted survival plasticity by inducing EMT and enhancing FA uptake via adenosine monophosphate-activated protein kinase activation. Notably, this desaturation reprogramming increased transforming growth factor-β2, effectively sustaining aggressive phenotypes and metabolic plasticity during EMT. CONCLUSIONS These findings revealed a metabolic reprogramming from SCD-dependent to FADS2-dependent desaturation during cancer EMT and progression, which concurrently supports EMT plasticity. Targeting desaturation reprogramming represents a potential vulnerability for cancer metabolic therapy.
Collapse
Affiliation(s)
- Zhicong Chen
- Department of Obstetrics and GynecologyCenter for Reproductive MedicineGuangdong Provincial Key Laboratory of Major Obstetric DiseasesGuangdong Provincial Clinical Research Center for Obstetrics and GynecologyGuangdong‐Hong Kong‐Macao Greater Bay Area Higher Education Joint Laboratory of Maternal‐Fetal MedicineThe Third Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongP. R. China
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Yanqing Gong
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Fukai Chen
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
| | - Hyeon Jeong Lee
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
- College of Biomedical Engineering & Instrument ScienceKey Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jinqin Qian
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Jing Zhao
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua UniversityBeijingP. R. China
| | - Wenpeng Zhang
- State Key Laboratory of Precision Measurement Technology and InstrumentsDepartment of Precision InstrumentTsinghua UniversityBeijingP. R. China
| | - Yamin Li
- Department of Biomedical EngineeringTufts UniversityMedfordMassachusettsUSA
| | - Yihui Zhou
- College of Biomedical Engineering & Instrument ScienceKey Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qiaobing Xu
- Department of Biomedical EngineeringTufts UniversityMedfordMassachusettsUSA
| | - Yu Xia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua UniversityBeijingP. R. China
| | - Liqun Zhou
- Department of UrologyPeking University First HospitalBeijingP. R. China
| | - Ji‐Xin Cheng
- Department of Biomedical EngineeringDepartment of Electrical and Computer EngineeringPhotonics CenterBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
5
|
Zhang W, Liu H, Zhu B, Li W, Han X, Fu J, Luo R, Wang H, Wang J. Advances in Cytosolic Delivery of Proteins: Approaches, Challenges, and Emerging Technologies. Chem Biodivers 2025:e202401713. [PMID: 39921680 DOI: 10.1002/cbdv.202401713] [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: 07/14/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Although therapeutic proteins have achieved recognized clinical success, they are inherently membrane impermeable, which limits them to acting only on extracellular or membrane-associated targets. Developing an efficient protein delivery method will provide a unique opportunity for intracellular target-related therapeutic proteins. In this review article, we summarize the different pathways by which cells take up proteins. These pathways fall into two main categories: One in which proteins are transported directly across the cell membrane and the other through endocytosis. At the same time, important features to ensure successful delivery through these pathways are highlighted. We then provide a comprehensive overview of the latest developments in the transduction of covalent protein modifications, such as coupling cell-penetrating motifs and supercharging, as well as the use of nanocarriers to mediate protein transport, such as liposomes, polymers, and inorganic nanoparticles. Finally, we emphasize the existing challenges of cytoplasmic protein delivery and provide an outlook for future progress.
Collapse
Affiliation(s)
- Wenyan Zhang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Huiling Liu
- Gansu Provincial Hospital, Lanzhou, Gansu, China
| | | | - Wen Li
- Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xue Han
- Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Jiaojiao Fu
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Renjie Luo
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Haiyan Wang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jinxia Wang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
6
|
Nguyen T, Gebo C, Lu J, Popoola DO, Thomas SJ, Li Y, Waickman AT. Development and optimization of an mRNA-vectored single-chain IgA1 isotype monoclonal antibody with potential to treat or prevent dengue virus infection. Antiviral Res 2025; 234:106078. [PMID: 39778815 DOI: 10.1016/j.antiviral.2025.106078] [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: 09/23/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025]
Abstract
Dengue virus (DENV) is a rapidly expanding infectious disease threat that causes an estimated 100 million symptomatic infections every year. A barrier to preventing DENV infections with traditional vaccines or prophylactic monoclonal antibody (mAb) therapies is the phenomenon of Antibody-Dependent Enhancement (ADE), wherein sub-neutralizing levels of DENV-specific IgG antibodies can enhance infection and pathogenesis rather than providing protection from disease. Fortunately, IgG is not the only antibody isotype capable of binding and neutralizing DENV, as DENV-specific IgA1 isotype mAbs can bind and neutralize DENV while without exhibiting any ADE activity. However, the development of IgA1-based mAb therapies is currently hindered by inefficient in vitro expression systems and the lack of saleable purification platforms. Accordingly, alternative delivery modalities are required to realize the therapeutic potential of IgA-based infectious-disease therapies. In this study we describe the development and optimization of a DENV-specific single-chain IgA construct that retains the desirable biological properties of the parental IgA mAb yet is compatible with efficient in vivo delivery with a novel/liver-tropic lipid nanoparticle. We propose that this platform is uniquely and exceptionally well suited for preventing and/or treating DENV infections and may have broad applicability in the greater infectious disease space in situations where the use of IgG isotype mAbs may be counterindicated.
Collapse
MESH Headings
- Dengue/prevention & control
- Dengue/therapy
- Dengue/drug therapy
- Dengue/immunology
- Dengue Virus/immunology
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/genetics
- Antibodies, Viral/therapeutic use
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Humans
- Immunoglobulin A/genetics
- Immunoglobulin A/immunology
- Immunoglobulin A/therapeutic use
- Antibody-Dependent Enhancement
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Neutralizing/genetics
- RNA, Messenger/genetics
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/therapeutic use
- Mice
- Nanoparticles
Collapse
Affiliation(s)
- Thanh Nguyen
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Chad Gebo
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Joseph Lu
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA; Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - David O Popoola
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Stephen J Thomas
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA; Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Yamin Li
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Adam T Waickman
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA; Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
7
|
Cheng J, Jian L, Chen Z, Li Z, Yu Y, Wu Y. In Vivo Delivery Processes and Development Strategies of Lipid Nanoparticles. Chembiochem 2024; 25:e202400481. [PMID: 39101874 DOI: 10.1002/cbic.202400481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/06/2024]
Abstract
Lipid nanoparticles (LNPs) represent an advanced and highly efficient delivery system for RNA molecules, demonstrating exceptional biocompatibility and remarkable delivery efficiency. This is evidenced by the clinical authorization of three LNP formulations: Patisiran, BNT162b2, and mRNA-1273. To further maximize the efficacy of RNA-based therapy, it is imperative to develop more potent LNP delivery systems that can effectively protect inherently unstable and negatively charged RNA molecules from degradation by nucleases, while facilitating their cellular uptake into target cells. Therefore, this review presents feasible strategies commonly employed for the development of efficient LNP delivery systems. The strategies encompass combinatorial chemistry for large-scale synthesis of ionizable lipids, rational design strategy of ionizable lipids, functional molecules-derived lipid molecules, the optimization of LNP formulations, and the adjustment of particle size and charge property of LNPs. Prior to introducing these developing strategies, in vivo delivery processes of LNPs, a crucial determinant influencing the clinical translation of LNP formulations, is described to better understand how to develop LNP delivery systems.
Collapse
Affiliation(s)
- Jiashun Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lina Jian
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhaolin Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhuoyuan Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaobang Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
8
|
Haghighi E, Abolmaali SS, Dehshahri A, Mousavi Shaegh SA, Azarpira N, Tamaddon AM. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. J Nanobiotechnology 2024; 22:710. [PMID: 39543630 PMCID: PMC11566655 DOI: 10.1186/s12951-024-02972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
RNA therapeutics, such as mRNA, siRNA, and CRISPR-Cas9, present exciting avenues for treating diverse diseases. However, their potential is commonly hindered by vulnerability to degradation and poor cellular uptake, requiring effective delivery systems. Lipid nanoparticles (LNPs) have emerged as a leading choice for in vivo RNA delivery, offering protection against degradation, enhanced cellular uptake, and facilitation of endosomal escape. However, LNPs encounter numerous challenges for targeted RNA delivery in vivo, demanding advanced particle engineering, surface functionalization with targeting ligands, and a profound comprehension of the biological milieu in which they function. This review explores the structural and physicochemical characteristics of LNPs, in-vivo fate, and customization for RNA therapeutics. We highlight the quality-by-design (QbD) approach for targeted delivery beyond the liver, focusing on biodistribution, immunogenicity, and toxicity. In addition, we explored the current challenges and strategies associated with LNPs for in-vivo RNA delivery, such as ensuring repeated-dose efficacy, safety, and tissue-specific gene delivery. Furthermore, we provide insights into the current clinical applications in various classes of diseases and finally prospects of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
- Elahe Haghighi
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Azarpira
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Lu RM, Hsu HE, Perez SJLP, Kumari M, Chen GH, Hong MH, Lin YS, Liu CH, Ko SH, Concio CAP, Su YJ, Chang YH, Li WS, Wu HC. Current landscape of mRNA technologies and delivery systems for new modality therapeutics. J Biomed Sci 2024; 31:89. [PMID: 39256822 PMCID: PMC11389359 DOI: 10.1186/s12929-024-01080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Realizing the immense clinical potential of mRNA-based drugs will require continued development of methods to safely deliver the bioactive agents with high efficiency and without triggering side effects. In this regard, lipid nanoparticles have been successfully utilized to improve mRNA delivery and protect the cargo from extracellular degradation. Encapsulation in lipid nanoparticles was an essential factor in the successful clinical application of mRNA vaccines, which conclusively demonstrated the technology's potential to yield approved medicines. In this review, we begin by describing current advances in mRNA modifications, design of novel lipids and development of lipid nanoparticle components for mRNA-based drugs. Then, we summarize key points pertaining to preclinical and clinical development of mRNA therapeutics. Finally, we cover topics related to targeted delivery systems, including endosomal escape and targeting of immune cells, tumors and organs for use with mRNA vaccines and new treatment modalities for human diseases.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Hsiang-En Hsu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Monika Kumari
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Guan-Hong Chen
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ming-Hsiang Hong
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Yin-Shiou Lin
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ching-Hang Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Yi-Jen Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Yi-Han Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Wen-Shan Li
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Chemistry, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| | - Han-Chung Wu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| |
Collapse
|
10
|
Wang J, Zhang S, Li Y, Xu Q, Kritzer JA. Investigating the Cytosolic Delivery of Proteins by Lipid Nanoparticles Using the Chloroalkane Penetration Assay. Biochemistry 2024:10.1021/acs.biochem.3c00614. [PMID: 38334719 PMCID: PMC11787851 DOI: 10.1021/acs.biochem.3c00614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Protein therapeutics are an expanding area for research and drug development, and lipid nanoparticles (LNPs) are the most prominent nonviral vehicles for protein delivery. The most common methods for assessing protein delivery by LNPs include assays that measure the total amount of protein taken up by cells and assays that measure the phenotypic changes associated with protein delivery. However, assays for total cellular uptake include large amounts of protein that are trapped in endosomes or are otherwise nonfunctional. Assays for functional delivery are important, but the readouts are indirect and amplified, limiting the quantitative interpretation. Here, we apply an assay for cytosolic delivery, the chloroalkane penetration assay (CAPA), to measure the cytosolic delivery of a (-30) green fluorescent protein (GFP) fused to Cre recombinase (Cre(-30)GFP) fusion protein by LNPs. We compare these data to the data from total cellular uptake and functional delivery assays to provide a richer analysis of uptake and endosomal escape for LNP-mediated protein delivery. We also use CAPA for a screen of a small library of lipidoids, identifying those with a promising ability to deliver Cre(-30)GFP to the cytosol of mammalian cells. With careful controls and optimized conditions, we expect that CAPA will be a useful tool for investigating the rate, efficiency, and mechanisms of LNP-mediated delivery of therapeutic proteins.
Collapse
Affiliation(s)
- Jing Wang
- Department of Chemistry, Tufts University, Medford, MA 02155
| | - Shiying Zhang
- Department of Chemistry, Tufts University, Medford, MA 02155
| | - Yamin Li
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA 02155
| | - Qiaobing Xu
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA 02155
| | | |
Collapse
|
11
|
Li T, Luo R, Su L, Lv F, Mei L, Yu Y. Advanced Materials and Delivery Systems for Enhancement of Chimeric Antigen Receptor Cells. SMALL METHODS 2023; 7:e2300880. [PMID: 37653606 DOI: 10.1002/smtd.202300880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/12/2023] [Indexed: 09/02/2023]
Abstract
Chimeric antigen receptor (CAR) cell therapy is a great success and breakthrough in immunotherapy. However, there are still lots of barriers to its wide use in clinical, including long time consumption, high cost, and failure against solid tumors. For these challenges, researches are deplored to explore CAR cells to more appliable products in clinical. This minireview focuses on the advanced non-viral materials for CAR-T transfection ex vivo with better performance, delivery systems combined with other therapy for enhancement of CAR-T therapy in solid tumors. In addition, the targeted delivery platform for CAR cells in vivo generation as a breakthrough technology as its low cost and convenience. In the end, the prospective direction and future of CAR cell therapy are discussed.
Collapse
Affiliation(s)
- Tingxuan Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Ran Luo
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Lina Su
- Department of Pharmacy, Qujing Medical College, Qujing, Yunnan, 655000, P. R. China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yongkang Yu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
12
|
Wang X, Li Y, Wang X, Sandoval DM, He Z, A S, Sáez IL, Wang W. Guanidyl-Rich Poly(β Amino Ester)s for Universal Functional Cytosolic Protein Delivery and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) Cas9 Ribonucleoprotein Based Gene Editing. ACS NANO 2023; 17:17799-17810. [PMID: 37669145 PMCID: PMC10540258 DOI: 10.1021/acsnano.3c03269] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023]
Abstract
Protein therapeutics are highly promising for complex disease treatment. However, the lack of ideal delivery vectors impedes their clinical use, especially the carriers for in vivo delivery of functional cytosolic protein. In this study, we modified poly(β amino ester)s (PAEs) with a phenyl guanidine (PG) group to enhance their suitability for cytosolic protein delivery. The effects of the PG group on protein binding, cell internalization, protein function protection, and endo/lysosomal escape were systematically evaluated. Compared to the unmodified PAEs (L3), guanidyl rich PAEs (L3PG) presented superior efficiency of protein binding and protein internalization, mainly via clathrin-mediated endocytosis. In addition, both PAEs showed robust capabilities to deliver cytosolic proteins with different molecular weight (ranging from 30 to 464 kDa) and isoelectric points (ranging from 4.3 to 9), which were significantly improved in comparison with the commercial reagents of PULsin and Pierce Protein Transection Reagent. Moreover, L3PG successfully delivered Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) Cas9 ribonucleoprotein (RNP) into HeLa cells expressing green fluorescent protein (GFP) and achieved more than 80% GFP expression knockout. These results demonstrated that guanidyl modification on PAEs can enhance its capabilities for intracellular delivery of cytosolic functional proteins and CRISPR/Cas9 ribonucleoprotein. The guanidyl-rich PAEs are promising nonviral vectors for functional protein delivery and potential use in protein and nuclease-based gene editing therapies.
Collapse
Affiliation(s)
- Xianqing Wang
- Charles
Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
- Research
and Clinical Translation Center of Gene Medicine and Tissue Engineering,
School of Public Health, Anhui University
of Science and Technology, Huainan 232001, China
| | - Yinghao Li
- Charles
Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Xi Wang
- Research
and Clinical Translation Center of Gene Medicine and Tissue Engineering,
School of Public Health, Anhui University
of Science and Technology, Huainan 232001, China
| | - Dario M. Sandoval
- Charles
Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Zhonglei He
- Charles
Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
- Research
and Clinical Translation Center of Gene Medicine and Tissue Engineering,
School of Public Health, Anhui University
of Science and Technology, Huainan 232001, China
| | - Sigen A
- Charles
Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
- Research
and Clinical Translation Center of Gene Medicine and Tissue Engineering,
School of Public Health, Anhui University
of Science and Technology, Huainan 232001, China
| | - Irene Lara Sáez
- Charles
Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Wenxin Wang
- Charles
Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
- Research
and Clinical Translation Center of Gene Medicine and Tissue Engineering,
School of Public Health, Anhui University
of Science and Technology, Huainan 232001, China
| |
Collapse
|
13
|
Ma H, Xing F, Zhou Y, Yu P, Luo R, Xu J, Xiang Z, Rommens PM, Duan X, Ritz U. Design and fabrication of intracellular therapeutic cargo delivery systems based on nanomaterials: current status and future perspectives. J Mater Chem B 2023; 11:7873-7912. [PMID: 37551112 DOI: 10.1039/d3tb01008b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Intracellular cargo delivery, the introduction of small molecules, proteins, and nucleic acids into a specific targeted site in a biological system, is an important strategy for deciphering cell function, directing cell fate, and reprogramming cell behavior. With the advancement of nanotechnology, many researchers use nanoparticles (NPs) to break through biological barriers to achieving efficient targeted delivery in biological systems, bringing a new way to realize efficient targeted drug delivery in biological systems. With a similar size to many biomolecules, NPs possess excellent physical and chemical properties and a certain targeting ability after functional modification on the surface of NPs. Currently, intracellular cargo delivery based on NPs has emerged as an important strategy for genome editing regimens and cell therapy. Although researchers can successfully deliver NPs into biological systems, many of them are delivered very inefficiently and are not specifically targeted. Hence, the development of efficient, target-capable, and safe nanoscale drug delivery systems to deliver therapeutic substances to cells or organs is a major challenge today. In this review, on the basis of describing the research overview and classification of NPs, we focused on the current research status of intracellular cargo delivery based on NPs in biological systems, and discuss the current problems and challenges in the delivery process of NPs in biological systems.
Collapse
Affiliation(s)
- Hong Ma
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Fei Xing
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Ludwigstraße 23, 35392 Giessen, Germany
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Rong Luo
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiawei Xu
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Xin Duan
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedic Surgery, The Fifth People's Hospital of Sichuan Province, Chengdu, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
14
|
Ye Z, Harmon J, Ni W, Li Y, Wich D, Xu Q. The mRNA Vaccine Revolution: COVID-19 Has Launched the Future of Vaccinology. ACS NANO 2023; 17:15231-15253. [PMID: 37535899 DOI: 10.1021/acsnano.2c12584] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
During the COVID-19 pandemic, mRNA (mRNA) vaccines emerged as leading vaccine candidates in a record time. Nonreplicating mRNA (NRM) and self-amplifying mRNA (SAM) technologies have been developed into high-performing and clinically viable vaccines against a range of infectious agents, notably SARS-CoV-2. mRNA vaccines demonstrate efficient in vivo delivery, long-lasting stability, and nonexistent risk of infection. The stability and translational efficiency of in vitro transcription (IVT)-mRNA can be further increased by modulating its structural elements. In this review, we present a comprehensive overview of the recent advances, key applications, and future challenges in the field of mRNA-based vaccinology.
Collapse
Affiliation(s)
- Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Joseph Harmon
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Wei Ni
- Department of Medical Oncology, Dana-Farber Cancer Institute at Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yamin Li
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York 13210, United States
| | - Douglas Wich
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
15
|
Zheng Q, Wang W, Zhou Y, Mo J, Chang X, Zha Z, Zha L. Synthetic nanoparticles for the delivery of CRISPR/Cas9 gene editing system: classification and biomedical applications. Biomater Sci 2023; 11:5361-5389. [PMID: 37381725 DOI: 10.1039/d3bm00788j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Gene editing has great potential in biomedical research including disease diagnosis and treatment. Clustered regularly interspaced short palindromic repeats (CRISPR) is the most straightforward and cost-effective method. The efficient and precise delivery of CRISPR can impact the specificity and efficacy of gene editing. In recent years, synthetic nanoparticles have been discovered as effective CRISPR/Cas9 delivery vehicles. We categorized synthetic nanoparticles for CRISPR/Cas9 delivery and discribed their advantages and disadvantages. Further, the building blocks of different kinds of nanoparticles and their applications in cells/tissues, cancer and other diseases were described in detail. Finally, the challenges encountered in the clinical application of CRISPR/Cas9 delivery materials were discussed, and potential solutions were provided regarding efficiency and biosafety issues.
Collapse
Affiliation(s)
- Qi Zheng
- International Immunology Centre, Anhui Agricultural University, Hefei 230036, P. R. China.
| | - Weitao Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China.
| | - Yuhang Zhou
- International Immunology Centre, Anhui Agricultural University, Hefei 230036, P. R. China.
| | - Jiayin Mo
- International Immunology Centre, Anhui Agricultural University, Hefei 230036, P. R. China.
| | - Xinyue Chang
- International Immunology Centre, Anhui Agricultural University, Hefei 230036, P. R. China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China.
| | - Lisha Zha
- International Immunology Centre, Anhui Agricultural University, Hefei 230036, P. R. China.
| |
Collapse
|
16
|
Li W, Shao L, Liu J, Sheng J, Zheng Q, Wang M. Intracellular delivery of bacterial effectors for cancer therapy using biodegradable lipid nanoparticles. Biomater Sci 2023; 11:3172-3179. [PMID: 36919841 DOI: 10.1039/d3bm00008g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Bacterial effector proteins are virulence factors that are secreted and mediate orthogonal post-translational modifications of proteins that are not found naturally in mammalian systems. They hold great promise for developing biotherapeutics by regulating malignant cell signaling in a specific and targeted manner. However, delivering bacterial effectors into disease cells poses a significant challenge to their therapeutic potential. In this study, we report on the design of a combinatorial library of bioreducible lipid nanoparticles containing disulfide bonds for highly efficient bacterial effector delivery and potential cancer therapy. A leading lipid, PPPDA-O16B, identified from the library, can encapsulate and deliver DNA plasmids into cells. The gene cargo is released in response to the reductive cellular environment that is upregulated in cancer cells, leading to enhanced gene delivery and protein expression efficiency. Furthermore, we demonstrate that PPPDA-O16B can deliver the bacterial effector protein, DUF5, to degrade mutant RAS and inactivate downstream MAPK signaling cascades to suppress cancer cell growth in vitro and in tumor-bearing mouse xenografts. This strategy of delivering bacterial effectors using biodegradable lipid nanoparticles can be expanded for cancer cell signaling regulation and antitumor studies.
Collapse
Affiliation(s)
- Wenting Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Leihou Shao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ji Liu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinhan Sheng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qizhen Zheng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Development of a Library of Disulfide Bond-Containing Cationic Lipids for mRNA Delivery. Pharmaceutics 2023; 15:pharmaceutics15020477. [PMID: 36839799 PMCID: PMC9961079 DOI: 10.3390/pharmaceutics15020477] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
Lipid nanoparticles (LNPs) are the commonly used delivery tools for messenger RNA (mRNA) therapy and play an indispensable role in the success of COVID-19 mRNA vaccines. Ionizable cationic lipids are the most important component in LNPs. Herein, we developed a series of new ionizable lipids featuring bioreducible disulfide bonds, and constructed a library of lipids derived from dimercaprol. LNPs prepared from these ionizable lipids could be stored at 4 °C for a long term and are non-toxic toward HepG2 and 293T cells. In vivo experiments demonstrated that the best C4S18A formulations, which embody linoleoyl tails, show strong firefly luciferase (Fluc) mRNA expression in the liver and spleen via intravenous (IV) injection, or at the local injection site via intramuscular injection (IM). The newly designed ionizable lipids can be potentially safe and high-efficiency nanomaterials for mRNA therapy.
Collapse
|
18
|
Allemailem KS, Alsahli MA, Almatroudi A, Alrumaihi F, Alkhaleefah FK, Rahmani AH, Khan AA. Current updates of CRISPR/Cas9-mediated genome editing and targeting within tumor cells: an innovative strategy of cancer management. Cancer Commun (Lond) 2022; 42:1257-1287. [PMID: 36209487 PMCID: PMC9759771 DOI: 10.1002/cac2.12366] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/19/2022] [Accepted: 09/21/2022] [Indexed: 01/25/2023] Open
Abstract
Clustered regularly interspaced short palindromic repeats-associated protein (CRISPR/Cas9), an adaptive microbial immune system, has been exploited as a robust, accurate, efficient and programmable method for genome targeting and editing. This innovative and revolutionary technique can play a significant role in animal modeling, in vivo genome therapy, engineered cell therapy, cancer diagnosis and treatment. The CRISPR/Cas9 endonuclease system targets a specific genomic locus by single guide RNA (sgRNA), forming a heteroduplex with target DNA. The Streptococcus pyogenes Cas9/sgRNA:DNA complex reveals a bilobed architecture with target recognition and nuclease lobes. CRISPR/Cas9 assembly can be hijacked, and its nanoformulation can be engineered as a delivery system for different clinical utilizations. However, the efficient and safe delivery of the CRISPR/Cas9 system to target tissues and cancer cells is very challenging, limiting its clinical utilization. Viral delivery strategies of this system may have many advantages, but disadvantages such as immune system stimulation, tumor promotion risk and small insertion size outweigh these advantages. Thus, there is a desperate need to develop an efficient non-viral physical delivery system based on simple nanoformulations. The delivery strategies of CRISPR/Cas9 by a nanoparticle-based system have shown tremendous potential, such as easy and large-scale production, combination therapy, large insertion size and efficient in vivo applications. This review aims to provide in-depth updates on Streptococcus pyogenic CRISPR/Cas9 structure and its mechanistic understanding. In addition, the advances in its nanoformulation-based delivery systems, including lipid-based, polymeric structures and rigid NPs coupled to special ligands such as aptamers, TAT peptides and cell-penetrating peptides, are discussed. Furthermore, the clinical applications in different cancers, clinical trials and future prospects of CRISPR/Cas9 delivery and genome targeting are also discussed.
Collapse
Affiliation(s)
- Khaled S. Allemailem
- Department of Medical Laboratories, College of Applied Medical SciencesQassim UniversityBuraydahSaudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical SciencesQassim UniversityBuraydahSaudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical SciencesQassim UniversityBuraydahSaudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical SciencesQassim UniversityBuraydahSaudi Arabia
| | | | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical SciencesQassim UniversityBuraydahSaudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health SciencesCollege of Applied Medical SciencesQassim UniversityBuraydahSaudi Arabia
| |
Collapse
|
19
|
Foley RA, Sims RA, Duggan EC, Olmedo JK, Ma R, Jonas SJ. Delivering the CRISPR/Cas9 system for engineering gene therapies: Recent cargo and delivery approaches for clinical translation. Front Bioeng Biotechnol 2022; 10:973326. [PMID: 36225598 PMCID: PMC9549251 DOI: 10.3389/fbioe.2022.973326] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022] Open
Abstract
Clustered Regularly Interspaced Short Palindromic Repeats associated protein 9 (CRISPR/Cas9) has transformed our ability to edit the human genome selectively. This technology has quickly become the most standardized and reproducible gene editing tool available. Catalyzing rapid advances in biomedical research and genetic engineering, the CRISPR/Cas9 system offers great potential to provide diagnostic and therapeutic options for the prevention and treatment of currently incurable single-gene and more complex human diseases. However, significant barriers to the clinical application of CRISPR/Cas9 remain. While in vitro, ex vivo, and in vivo gene editing has been demonstrated extensively in a laboratory setting, the translation to clinical studies is currently limited by shortfalls in the precision, scalability, and efficiency of delivering CRISPR/Cas9-associated reagents to their intended therapeutic targets. To overcome these challenges, recent advancements manipulate both the delivery cargo and vehicles used to transport CRISPR/Cas9 reagents. With the choice of cargo informing the delivery vehicle, both must be optimized for precision and efficiency. This review aims to summarize current bioengineering approaches to applying CRISPR/Cas9 gene editing tools towards the development of emerging cellular therapeutics, focusing on its two main engineerable components: the delivery vehicle and the gene editing cargo it carries. The contemporary barriers to biomedical applications are discussed within the context of key considerations to be made in the optimization of CRISPR/Cas9 for widespread clinical translation.
Collapse
Affiliation(s)
- Ruth A. Foley
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
- Department of Bioengineering, University of California, Los Angeles, CA, United States
| | - Ruby A. Sims
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
- California NanoSystems Institute, University of California, Los Angeles, CA, United States
| | - Emily C. Duggan
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - Jessica K. Olmedo
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - Rachel Ma
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
| | - Steven J. Jonas
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
- California NanoSystems Institute, University of California, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States
| |
Collapse
|
20
|
Yamada Y, Sato Y, Nakamura T, Harashima H. Innovative cancer nanomedicine based on immunology, gene editing, intracellular trafficking control. J Control Release 2022; 348:357-369. [PMID: 35623492 DOI: 10.1016/j.jconrel.2022.05.033] [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: 03/25/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022]
Abstract
The recent rapid progress in the area of drug delivery systems (DDS) has opened a new era in medicine with a strong linkage to understanding the molecular mechanisms associated with cancer survival. In this review, we summarize new cancer strategies that have recently been developed based on our DDS technology. Cancer immunotherapy will be improved based on the concept of the cancer immunity cycle, which focuses on dynamic interactions between various types of cancer and immune cells in our body. The new technology of genome editing will also be discussed with reference to how these new DDS technologies can be used to introduce therapeutic cargoes into our body. Lastly, a new organelle, mitochondria will be the focus of creating a new cancer treatment strategy by a MITO-Porter which can deliver macromolecules directly to mitochondria of cancer cells via a membrane fusion approach and the impact of controlled intracellular trafficking will be discussed.
Collapse
Affiliation(s)
- Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Japan Science and Technology Agency (JST) Fusion Oriented REsearch for disruptive Science and Technology (FOREST) Program, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
21
|
Li Y, Ye Z, Yang H, Xu Q. Tailoring combinatorial lipid nanoparticles for intracellular delivery of nucleic acids, proteins, and drugs. Acta Pharm Sin B 2022; 12:2624-2639. [PMID: 35755280 PMCID: PMC9214058 DOI: 10.1016/j.apsb.2022.04.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/17/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
Lipid nanoparticle (LNP)-based drug delivery systems have become the most clinically advanced non-viral delivery technology. LNPs can encapsulate and deliver a wide variety of bioactive agents, including the small molecule drugs, proteins and peptides, and nucleic acids. However, as the physicochemical properties of small- and macromolecular cargos can vary drastically, every LNP carrier system needs to be carefully tailored in order to deliver the cargo molecules in a safe and efficient manner. Our group applied the combinatorial library synthesis approach and in vitro and in vivo screening strategy for the development of LNP delivery systems for drug delivery. In this Review, we highlight our recent progress in the design, synthesis, characterization, evaluation, and optimization of combinatorial LNPs with novel structures and properties for the delivery of small- and macromolecular therapeutics both in vitro and in vivo. These delivery systems have enormous potentials for cancer therapy, antimicrobial applications, gene silencing, genome editing, and more. We also discuss the key challenges to the mechanistic study and clinical translation of new LNP-enabled therapeutics.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Hanyi Yang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
22
|
Xu Z, Wang Q, Zhong H, Jiang Y, Shi X, Yuan B, Yu N, Zhang S, Yuan X, Guo S, Yang Y. Carrier strategies boost the application of CRISPR/Cas system in gene therapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20210081. [PMID: 37323878 PMCID: PMC10190933 DOI: 10.1002/exp.20210081] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023]
Abstract
Emerging clustered regularly interspaced short palindromic repeat/associated protein (CRISPR/Cas) genome editing technology shows great potential in gene therapy. However, proteins and nucleic acids suffer from enzymatic degradation in the physiological environment and low permeability into cells. Exploiting carriers to protect the CRISPR system from degradation, enhance its targeting of specific tissues and cells, and reduce its immunogenicity is essential to stimulate its clinical applications. Here, the authors review the state-of-the-art CRISPR delivery systems and their applications, and describe strategies to improve the safety and efficacy of CRISPR mediated genome editing, categorized by three types of cargo formats, that is, Cas: single-guide RNA ribonucleoprotein, Cas mRNA and single-guide RNA, and Cas plasmid expressing CRISPR/Cas systems. The authors hope this review will help develop safe and efficient nanomaterial-based carriers for CRISPR tools.
Collapse
Affiliation(s)
- Zunkai Xu
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation CenterChengduChina
| | - Haiping Zhong
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Yaoyao Jiang
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Xiaoguang Shi
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Bo Yuan
- School of MedicineNankai UniversityTianjinChina
- Tianjin Key Laboratory of Ophthalmology and Visual ScienceTianjin Eye InstituteTianjin Eye HospitalTianjinChina
| | - Na Yu
- Translational Medicine CenterKey Laboratory of Molecular Target & Clinical PharmacologySchool of Pharmaceutical Sciences and The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of EducationDalian Minzu UniversityDalianChina
| | - Xiaoyong Yuan
- Tianjin Key Laboratory of Ophthalmology and Visual ScienceTianjin Eye InstituteTianjin Eye HospitalTianjinChina
- Clinical College of OphthalmologyTianjin Medical UniversityTianjinChina
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation CenterChengduChina
| |
Collapse
|
23
|
Feng S, Wang Z, Li A, Xie X, Liu J, Li S, Li Y, Wang B, Hu L, Yang L, Guo T. Strategies for High-Efficiency Mutation Using the CRISPR/Cas System. Front Cell Dev Biol 2022; 9:803252. [PMID: 35198566 PMCID: PMC8860194 DOI: 10.3389/fcell.2021.803252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated systems have revolutionized traditional gene-editing tools and are a significant tool for ameliorating gene defects. Characterized by high target specificity, extraordinary efficiency, and cost-effectiveness, CRISPR/Cas systems have displayed tremendous potential for genetic manipulation in almost any organism and cell type. Despite their numerous advantages, however, CRISPR/Cas systems have some inherent limitations, such as off-target effects, unsatisfactory efficiency of delivery, and unwanted adverse effects, thereby resulting in a desire to explore approaches to address these issues. Strategies for improving the efficiency of CRISPR/Cas-induced mutations, such as reducing off-target effects, improving the design and modification of sgRNA, optimizing the editing time and the temperature, choice of delivery system, and enrichment of sgRNA, are comprehensively described in this review. Additionally, several newly emerging approaches, including the use of Cas variants, anti-CRISPR proteins, and mutant enrichment, are discussed in detail. Furthermore, the authors provide a deep analysis of the current challenges in the utilization of CRISPR/Cas systems and the future applications of CRISPR/Cas systems in various scenarios. This review not only serves as a reference for improving the maturity of CRISPR/Cas systems but also supplies practical guidance for expanding the applicability of this technology.
Collapse
Affiliation(s)
- Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zilong Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Aifang Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xin Xie
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Junjie Liu
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Shuxuan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yalan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Baiyan Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lina Hu
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lianhe Yang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
24
|
Ye Z, Chen J, Zhao X, Li Y, Harmon J, Huang C, Chen J, Xu Q. In Vitro Engineering Chimeric Antigen Receptor Macrophages and T Cells by Lipid Nanoparticle-Mediated mRNA Delivery. ACS Biomater Sci Eng 2022; 8:722-733. [PMID: 35104103 DOI: 10.1021/acsbiomaterials.1c01532] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Chimeric antigen receptor (CAR)-engineered adoptive cell therapy marks a revolution in cancer treatment based on the highly successful responses to CAR T cell therapy in the treatment of blood cancers. Due to the versatile structure of CARs, this technology can be easily adapted to other immune cell types, including macrophages and NKs, and applied in the treatment of many other cancers. However, high costs and fatal adverse effects represent significant concerns for future development. In vitro transcribed (IVT) mRNA therapeutics, which possess a high safety profile and straightforward production methods, could provide a useful alternative for CAR cell construction. However, the low stability and transfection efficiency of IVT-mRNA in immune cells limit further applications. In this work, we successfully engineered CAR macrophages (CAR-Ms) and CAR T cells with CAR mRNA using lipid nanoparticles (LNPs). Both the LNP formulations and mRNA modifications were optimized for in vitro mRNA transfection. More importantly, the CAR macrophages and CAR T cells both demonstrated significant cytotoxic effects on B lymphoma in vitro, underscoring the great potential of mRNA-engineered adoptive cell therapy.
Collapse
Affiliation(s)
- Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Xuewei Zhao
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Joseph Harmon
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Changfeng Huang
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02139, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
25
|
Horn JM, Obermeyer AC. Genetic and Covalent Protein Modification Strategies to Facilitate Intracellular Delivery. Biomacromolecules 2021; 22:4883-4904. [PMID: 34855385 PMCID: PMC9310055 DOI: 10.1021/acs.biomac.1c00745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein-based therapeutics represent a rapidly growing segment of approved disease treatments. Successful intracellular delivery of proteins is an important precondition for expanded in vivo and in vitro applications of protein therapeutics. Direct modification of proteins and peptides for improved cytosolic translocation are a promising method of increasing delivery efficiency and expanding the viability of intracellular protein therapeutics. In this Review, we present recent advances in both synthetic and genetic protein modifications for intracellular delivery. Active endocytosis-based and passive internalization pathways are discussed, followed by a review of modification methods for improved cytosolic delivery. After establishing how proteins can be modified, general strategies for facilitating intracellular delivery, such as chemical supercharging or inclusion of cell-penetrating motifs, are covered. We then outline protein modifications that promote endosomal escape. We finally examine the delivery of two potential classes of therapeutic proteins, antibodies and associated antibody fragments, and gene editing proteins, such as cas9.
Collapse
|
26
|
Hershman RL, Li Y, Ma F, Xu Q, Van Deventer J. Intracellular Delivery of Antibodies for Selective Cell Signaling Interference. ChemMedChem 2021; 17:e202100678. [PMID: 34890114 DOI: 10.1002/cmdc.202100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Indexed: 11/11/2022]
Abstract
Many intracellular signaling events remain poorly characterized due to a general lack of tools to interfere with "undruggable" targets. Antibodies have the potential to elucidate intracellular mechanisms via targeted disruption of cell signaling cascades because of their ability to bind to a target with high specificity and affinity. However, due to their size and chemical composition, antibodies cannot innately cross the cell membrane, and thus access to the cytosol with these macromolecules has been limited. Here, we describe strategies for accessing the intracellular space with recombinant antibodies mediated by cationic lipid nanoparticles to selectively disrupt intracellular signaling events. Together, our results demonstrate the use of recombinantly produced antibodies, delivered at concentrations of 10 nM, to selectively interfere with signaling driven by a single posttranslational modification. Efficient intracellular delivery of engineered antibodies opens up possibilities for modulation of previously "undruggable" targets, including for potential therapeutic applications.
Collapse
Affiliation(s)
| | - Yamin Li
- Tufts University, Biomedical Engineering, UNITED STATES
| | - Feihe Ma
- Tufts University, Biomedical Engineering, UNITED STATES
| | - Qioabing Xu
- Tufts University, Biomedical Engineering, UNITED STATES
| | - James Van Deventer
- Tufts University, Chemical and Biological Engineering, 4 Colby St. Room 148, 02155, Medford, UNITED STATES
| |
Collapse
|
27
|
Considerations for the delivery of STING ligands in cancer immunotherapy. J Control Release 2021; 339:235-247. [PMID: 34592386 DOI: 10.1016/j.jconrel.2021.09.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/23/2022]
Abstract
Several studies have shown the importance of the cGAS-STING pathway in antigen-presenting cells for anti-cancer immunity. Cyclic GMP-AMP (cGAMP) - STING ligand is a negatively charged dinucleotide prone to degradation by hydrolases. Once administered in its soluble form, high doses are needed which in turn may cause side effects such as T cell apoptosis. Moreover, due to its negative charge, transfection of cGAMP into negatively-charged membrane cells is hampered. In order to achieve successful transfection and protection from enzymatic degradation there is a need for a suitable carrier for cGAMP. In this review, we therefore describe currently reported carriers for cGAMP, and correlate their characteristics to the effect they cause. To achieve targeted delivery to the tumor microenvironment, the route of administration and physicochemical parameters of the particles (containing a carrier and cGAMP) such as size and charge need to be determined. Therefore, the choice of the particle formulation and its impact on the preclinical outcome will be discussed.
Collapse
|
28
|
Qiu M, Li Y, Bloomer H, Xu Q. Developing Biodegradable Lipid Nanoparticles for Intracellular mRNA Delivery and Genome Editing. Acc Chem Res 2021; 54:4001-4011. [PMID: 34668716 DOI: 10.1021/acs.accounts.1c00500] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the U.S. Food and Drug Administration (FDA) granted emergency use authorization for two mRNA vaccines against SARS-CoV-2, mRNA-based technology has attracted broad attention from the scientific community to investors. When delivered intracellularly, mRNA has the ability to produce various therapeutic proteins, enabling the treatment of a variety of illnesses, including but not limited to infectious diseases, cancers, and genetic diseases. Accordingly, mRNA holds significant therapeutic potential and provides a promising means to target historically hard-to-treat diseases. Current clinical efforts harnessing mRNA-based technology are focused on vaccination, cancer immunotherapy, protein replacement therapy, and genome editing. The clinical translation of mRNA-based technology has been made possible by leveraging nanoparticle delivery methods. However, the application of mRNA for therapeutic purposes is still challenged by the need for specific, efficient, and safe delivery systems.This Account highlights key advances in designing and developing combinatorial synthetic lipid nanoparticles (LNPs) with distinct chemical structures and properties for in vitro and in vivo intracellular mRNA delivery. LNPs represent the most advanced nonviral nanoparticle delivery systems that have been extensively investigated for nucleic acid delivery. The aforementioned COVID-19 mRNA vaccines and one LNP-based small interfering RNA (siRNA) drug (ONPATTRO) have received clinical approval from the FDA, highlighting the success of synthetic ionizable lipids for in vivo nucleic acid delivery. In this Account, we first summarize the research efforts from our group on the development of bioreducible and biodegradable LNPs by leveraging the combinatorial chemistry strategy, such as the Michael addition reaction, which allows us to easily generate a large set of lipidoids with diverse chemical structures. Next, we discuss the utilization of a library screening strategy to identify optimal LNPs for targeted mRNA delivery and showcase the applications of the optimized LNPs in cell engineering and genome editing. Finally, we outline key challenges to the clinical translation of mRNA-based therapies and propose an outlook for future directions of the chemical design and optimization of LNPs to improve the safety and specificity of mRNA drugs. We hope this Account provides insight into the rational design of LNPs for facilitating the development of mRNA therapeutics, a transformative technology that promises to revolutionize future medicine.
Collapse
Affiliation(s)
- Min Qiu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Hanan Bloomer
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
- School of Medicine & Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts 02111, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
29
|
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
|
30
|
Le Saux S, Aubert-Pouëssel A, Mohamed KE, Martineau P, Guglielmi L, Devoisselle JM, Legrand P, Chopineau J, Morille M. Interest of extracellular vesicles in regards to lipid nanoparticle based systems for intracellular protein delivery. Adv Drug Deliv Rev 2021; 176:113837. [PMID: 34144089 DOI: 10.1016/j.addr.2021.113837] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/04/2021] [Accepted: 06/12/2021] [Indexed: 12/14/2022]
Abstract
Compared to chemicals that continue to dominate the overall pharmaceutical market, protein therapeutics offer the advantages of higher specificity, greater activity, and reduced toxicity. While nearly all existing therapeutic proteins were developed against soluble or extracellular targets, the ability for proteins to enter cells and target intracellular compartments can significantly broaden their utility for a myriad of exiting targets. Given their physical, chemical, biological instability that could induce adverse effects, and their limited ability to cross cell membranes, delivery systems are required to fully reveal their biological potential. In this context, as natural protein nanocarriers, extracellular vesicles (EVs) hold great promise. Nevertheless, if not present naturally, bringing an interest protein into EV is not an easy task. In this review, we will explore methods used to load extrinsic protein into EVs and compare these natural vectors to their close synthetic counterparts, liposomes/lipid nanoparticles, to induce intracellular protein delivery.
Collapse
|
31
|
Tomé I, Francisco V, Fernandes H, Ferreira L. High-throughput screening of nanoparticles in drug delivery. APL Bioeng 2021; 5:031511. [PMID: 34476328 PMCID: PMC8397474 DOI: 10.1063/5.0057204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022] Open
Abstract
The use of pharmacologically active compounds to manage and treat diseases is of utmost relevance in clinical practice. It is well recognized that spatial-temporal control over the delivery of these biomolecules will greatly impact their pharmacokinetic profile and ultimately their therapeutic effect. Nanoparticles (NPs) prepared from different materials have been tested successfully in the clinic for the delivery of several biomolecules including non-coding RNAs (siRNA and miRNA) and mRNAs. Indeed, the recent success of mRNA vaccines is in part due to progress in the delivery systems (NP based) that have been developed for many years. In most cases, the identification of the best formulation was done by testing a small number of novel formulations or by modification of pre-existing ones. Unfortunately, this is a low throughput and time-consuming process that hinders the identification of formulations with the highest potential. Alternatively, high-throughput combinatorial design of NP libraries may allow the rapid identification of formulations with the required release and cell/tissue targeting profile for a given application. Combinatorial approaches offer several advantages over conventional methods since they allow the incorporation of multiple components with varied chemical properties into materials, such as polymers or lipid-like materials, that will subsequently form NPs by self-assembly or chemical conjugation processes. The current review highlights the impact of high-throughput in the development of more efficient drug delivery systems with enhanced targeting and release kinetics. It also describes the current challenges in this research area as well as future directions.
Collapse
Affiliation(s)
| | - Vitor Francisco
- Biomaterials and Stem-Cell Based Therapeutics Group, Centre of Neuroscience and Cell Biology, University of Coimbra, 3060-197 Cantanhede, Portugal
| | | | - Lino Ferreira
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
32
|
Ho TC, Kim HS, Chen Y, Li Y, LaMere MW, Chen C, Wang H, Gong J, Palumbo CD, Ashton JM, Kim HW, Xu Q, Becker MW, Leong KW. Scaffold-mediated CRISPR-Cas9 delivery system for acute myeloid leukemia therapy. SCIENCE ADVANCES 2021; 7:eabg3217. [PMID: 34138728 PMCID: PMC8133753 DOI: 10.1126/sciadv.abg3217] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/23/2021] [Indexed: 05/06/2023]
Abstract
Leukemia stem cells (LSCs) sustain the disease and contribute to relapse in acute myeloid leukemia (AML). Therapies that ablate LSCs may increase the chance of eliminating this cancer in patients. To this end, we used a bioreducible lipidoid-encapsulated Cas9/single guide RNA (sgRNA) ribonucleoprotein [lipidoid nanoparticle (LNP)-Cas9 RNP] to target the critical gene interleukin-1 receptor accessory protein (IL1RAP) in human LSCs. To enhance LSC targeting, we loaded LNP-Cas9 RNP and the chemokine CXCL12α onto mesenchymal stem cell membrane-coated nanofibril (MSCM-NF) scaffolds mimicking the bone marrow microenvironment. In vitro, CXCL12α release induced migration of LSCs to the scaffolds, and LNP-Cas9 RNP induced efficient gene editing. IL1RAP knockout reduced LSC colony-forming capacity and leukemic burden. Scaffold-based delivery increased the retention time of LNP-Cas9 in the bone marrow cavity. Overall, sustained local delivery of Cas9/IL1RAP sgRNA via CXCL12α-loaded LNP/MSCM-NF scaffolds provides an effective strategy for attenuating LSC growth to improve AML therapy.
Collapse
Affiliation(s)
- Tzu-Chieh Ho
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Hye Sung Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Yumei Chen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | - Mark W LaMere
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Caroline Chen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Hui Wang
- Humanized Mouse Core Facility, Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
| | - Jing Gong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Cal D Palumbo
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Genomics Research Center, University of Rochester, Rochester, NY, USA
| | - John M Ashton
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Genomics Research Center, University of Rochester, Rochester, NY, USA
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, Republic of Korea
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | - Michael W Becker
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| |
Collapse
|
33
|
Chen J, Qiu M, Ye Z, Nyalile T, Li Y, Glass Z, Zhao X, Yang L, Chen J, Xu Q. In situ cancer vaccination using lipidoid nanoparticles. SCIENCE ADVANCES 2021; 7:7/19/eabf1244. [PMID: 33952519 PMCID: PMC8099179 DOI: 10.1126/sciadv.abf1244] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/17/2021] [Indexed: 05/08/2023]
Abstract
In situ vaccination is a promising strategy for cancer immunotherapy owing to its convenience and the ability to induce numerous tumor antigens. However, the advancement of in situ vaccination techniques has been hindered by low cross-presentation of tumor antigens and the immunosuppressive tumor microenvironment. To balance the safety and efficacy of in situ vaccination, we designed a lipidoid nanoparticle (LNP) to achieve simultaneously enhancing cross-presentation and STING activation. From combinatorial library screening, we identified 93-O17S-F, which promotes both the cross-presentation of tumor antigens and the intracellular delivery of cGAMP (STING agonist). Intratumor injection of 93-O17S-F/cGAMP in combination with pretreatment with doxorubicin exhibited excellent antitumor efficacy, with 35% of mice exhibiting total recovery from a primary B16F10 tumor and 71% of mice with a complete recovery from a subsequent challenge, indicating the induction of an immune memory against the tumor. This study provides a promising strategy for in situ cancer vaccination.
Collapse
Affiliation(s)
- Jinjin Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Min Qiu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Thomas Nyalile
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Zachary Glass
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Xuewei Zhao
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Liu Yang
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
34
|
Le Saux S, Aubert‐Pouëssel A, Ouchait L, Mohamed KE, Martineau P, Guglielmi L, Devoisselle J, Legrand P, Chopineau J, Morille M. Nanotechnologies for Intracellular Protein Delivery: Recent Progress in Inorganic and Organic Nanocarriers. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sarah Le Saux
- ICGM Universite Montpellier ENSCM, CNRS Montpellier France
| | | | - Lyria Ouchait
- ICGM Universite Montpellier ENSCM, CNRS Montpellier France
| | | | | | | | | | | | - Joël Chopineau
- ICGM Universite Montpellier ENSCM, CNRS Montpellier France
| | - Marie Morille
- ICGM Universite Montpellier ENSCM, CNRS Montpellier France
| |
Collapse
|
35
|
Zhao X, Glass Z, Chen J, Yang L, Kaplan DL, Xu Q. mRNA Delivery Using Bioreducible Lipidoid Nanoparticles Facilitates Neural Differentiation of Human Mesenchymal Stem Cells. Adv Healthc Mater 2021; 10:e2000938. [PMID: 32815325 DOI: 10.1002/adhm.202000938] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/12/2020] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in regenerative medicine and tissue engineering and delivering biological molecules into MSCs has been used to control stem cell behavior. However, the efficient delivery of large biomolecules such as DNA, RNA, and proteins into MSCs using nonviral delivery strategies remains an ongoing challenge. Herein, nanoparticles composed of cationic bioreducible lipid-like materials (lipidoids) are developed to intracellularly deliver mRNA into human mesenchymal stem cells (hMSCs). The delivery efficacy to hMSCs is improved by adding three excipients including cholesterol, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-polyethylene glycol (DSPE-PEG) during lipidoid nanoparticle formulation. Using an optimized lipidoid formulation, Cas9 mRNA and single guide RNA (sgRNA) targeting neuron restrictive silencing factor (NRSF) are delivered to hMSCs, leading to successful neural-like differentiation as demonstrated by the expression of synaptophysin (SYP), brain-derived neurotrophic factor (BDNF), neuron-specific enolase (NSE), and neuron-specific growth-associated protein (SCG10). Overall, a synthetic lipid formulation that can efficiently deliver mRNA to hMSCs is identified, leading to CRISPR-based gene knockdown to facilitate hMSCs transdifferentiation into neural-like lineage.
Collapse
Affiliation(s)
- Xuewei Zhao
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Liu Yang
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - David L. Kaplan
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| |
Collapse
|
36
|
Nanomaterials for Protein Delivery in Anticancer Applications. Pharmaceutics 2021; 13:pharmaceutics13020155. [PMID: 33503889 PMCID: PMC7910976 DOI: 10.3390/pharmaceutics13020155] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 12/16/2022] Open
Abstract
Nanotechnology platforms, such as nanoparticles, liposomes, dendrimers, and micelles have been studied extensively for various drug deliveries, to treat or prevent diseases by modulating physiological or pathological processes. The delivery drug molecules range from traditional small molecules to recently developed biologics, such as proteins, peptides, and nucleic acids. Among them, proteins have shown a series of advantages and potential in various therapeutic applications, such as introducing therapeutic proteins due to genetic defects, or used as nanocarriers for anticancer agents to decelerate tumor growth or control metastasis. This review discusses the existing nanoparticle delivery systems, introducing design strategies, advantages of using each system, and possible limitations. Moreover, we will examine the intracellular delivery of different protein therapeutics, such as antibodies, antigens, and gene editing proteins into the host cells to achieve anticancer effects and cancer vaccines. Finally, we explore the current applications of protein delivery in anticancer treatments.
Collapse
|
37
|
Tang H, Zhao X, Jiang X. Synthetic multi-layer nanoparticles for CRISPR-Cas9 genome editing. Adv Drug Deliv Rev 2021; 168:55-78. [PMID: 32147450 DOI: 10.1016/j.addr.2020.03.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/23/2020] [Accepted: 03/04/2020] [Indexed: 12/14/2022]
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR) has great potential to revolutionize biomedical research and disease therapy. The specific and efficient genome editing strongly depends on high efficiency of delivery of the CRISPR payloads. However, optimization of CRISPR delivery vehicles still remains a major obstacle. Recently, various non-viral vectors have been utilized to deliver CRISPR tools. Many of these vectors have multi-layer structures assembled. In this review, we will introduce the development of CRISPR-Cas9 systems and their general therapeutic applications by summarizing current CRISPR-Cas9 based clinical trials. We will highlight the multi-layer nanoparticles (NPs) that have been developed to deliver CRISPR cargos in vitro and in vivo for various purposes, as well the potential building blocks of multi-layer NPs. We will also discuss the challenges in making the CRISPR tools into viable pharmaceutical products and provide potential solutions on efficiency and biosafety issues.
Collapse
|
38
|
Li J, Røise JJ, He M, Das R, Murthy N. Non-viral strategies for delivering genome editing enzymes. Adv Drug Deliv Rev 2021; 168:99-117. [PMID: 32931860 DOI: 10.1016/j.addr.2020.09.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 08/02/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022]
Abstract
Genome-editing tools such as Cre recombinase (Cre), zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and most recently the clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein system have revolutionized biomedical research, agriculture, microbial engineering, and therapeutic development. Direct delivery of genome editing enzymes, as opposed to their corresponding DNA and mRNA precursors, is advantageous since they do not require transcription and/or translation. In addition, prolonged overexpression is a problem when delivering viral vector or plasmid DNA which is bypassed when delivering whole proteins. This lowers the risk of insertional mutagenesis and makes for relatively easier manufacturing. However, a major limitation of utilizing genome editing proteins in vivo is their low delivery efficiency, and currently the most successful strategy involves using potentially immunogenic viral vectors. This lack of safe and effective non-viral delivery systems is still a big hurdle for the clinical translation of such enzymes. This review discusses the challenges of non-viral delivery strategies of widely used genome editing enzymes, including Cre recombinase, ZFNs and TALENs, CRISPR/Cas9, and Cas12a (Cpf1) in their protein format and highlights recent innovations of non-viral delivery strategies which have the potential to overcome current delivery limitations and advance the clinical translation of genome editing.
Collapse
|
39
|
Zhang S, Shen J, Li D, Cheng Y. Strategies in the delivery of Cas9 ribonucleoprotein for CRISPR/Cas9 genome editing. Theranostics 2021; 11:614-648. [PMID: 33391496 PMCID: PMC7738854 DOI: 10.7150/thno.47007] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/31/2020] [Indexed: 12/26/2022] Open
Abstract
CRISPR/Cas9 genome editing has gained rapidly increasing attentions in recent years, however, the translation of this biotechnology into therapy has been hindered by efficient delivery of CRISPR/Cas9 materials into target cells. Direct delivery of CRISPR/Cas9 system as a ribonucleoprotein (RNP) complex consisting of Cas9 protein and single guide RNA (sgRNA) has emerged as a powerful and widespread method for genome editing due to its advantages of transient genome editing and reduced off-target effects. In this review, we summarized the current Cas9 RNP delivery systems including physical approaches and synthetic carriers. The mechanisms and beneficial roles of these strategies in intracellular Cas9 RNP delivery were reviewed. Examples in the development of stimuli-responsive and targeted carriers for RNP delivery are highlighted. Finally, the challenges of current Cas9 RNP delivery systems and perspectives in rational design of next generation materials for this promising field will be discussed.
Collapse
Affiliation(s)
- Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jiangtao Shen
- The Second People's Hospital of Taizhou affiliated to Yangzhou University, Taizhou, 225500, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| |
Collapse
|
40
|
Kumar R, Le N, Tan Z, Brown ME, Jiang S, Reineke TM. Efficient Polymer-Mediated Delivery of Gene-Editing Ribonucleoprotein Payloads through Combinatorial Design, Parallelized Experimentation, and Machine Learning. ACS NANO 2020; 14:17626-17639. [PMID: 33225680 DOI: 10.1021/acsnano.0c08549] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Chemically defined vectors such as cationic polymers are versatile alternatives to engineered viruses for the delivery of genome-editing payloads. However, their clinical translation hinges on rapidly exploring vast chemical design spaces and deriving structure-function relationships governing delivery performance. Here, we discovered a polymer for efficient intracellular ribonucleoprotein (RNP) delivery through combinatorial polymer design and parallelized experimental workflows. A chemically diverse library of 43 statistical copolymers was synthesized via combinatorial RAFT polymerization, realizing systematic variations in physicochemical properties. We selected cationic monomers that varied in their pKa values (8.1-9.2), steric bulk, and lipophilicity of their alkyl substituents. Co-monomers of varying hydrophilicity were also incorporated, enabling elucidation of the roles of protonation equilibria and hydrophobic-hydrophilic balance in vehicular properties and performance. We screened our multiparametric vector library through image cytometry and rapidly uncovered a hit polymer (P38), which outperforms state-of-the-art commercial transfection reagents, achieving nearly 60% editing efficiency via nonhomologous end-joining. Structure-function correlations underlying editing efficiency, cellular toxicity, and RNP uptake were probed through machine learning approaches to uncover the physicochemical basis of P38's performance. Although cellular toxicity and RNP uptake were solely determined by polyplex size distribution and protonation degree, respectively, these two polyplex design parameters were found to be inconsequential for enhancing editing efficiency. Instead, polymer hydrophobicity and the Hill coefficient, a parameter describing cooperativity-enhanced polymer deprotonation, were identified as the critical determinants of editing efficiency. Combinatorial synthesis and high-throughput characterization methodologies coupled with data science approaches enabled the rapid discovery of a polymeric vehicle that would have otherwise remained inaccessible to chemical intuition. The statistically derived design rules elucidated herein will guide the synthesis and optimization of future polymer libraries tailored for therapeutic applications of RNP-based genome editing.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ngoc Le
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zhe Tan
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary E Brown
- University Imaging Centers, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Shan Jiang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
41
|
Suzuki Y, Onuma H, Sato R, Sato Y, Hashiba A, Maeki M, Tokeshi M, Kayesh MEH, Kohara M, Tsukiyama-Kohara K, Harashima H. Lipid nanoparticles loaded with ribonucleoprotein-oligonucleotide complexes synthesized using a microfluidic device exhibit robust genome editing and hepatitis B virus inhibition. J Control Release 2020; 330:61-71. [PMID: 33333121 DOI: 10.1016/j.jconrel.2020.12.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/25/2022]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) system has considerable therapeutic potential for use in treating a wide range of intractable genetic and infectious diseases including hepatitis B virus (HBV) infections. While non-viral delivery technologies for the CRISPR/Cas system are expected to have clinical applications, difficulties associated with the clinically relevant synthesis of formulations and the poor efficiency of delivery severely hinder therapeutic genome editing. We report herein on the production of a lipid nanoparticle (LNP)-based CRISPR/Cas ribonucleoprotein (RNP) delivery nanoplatform synthesized using a clinically relevant mixer-equipped microfluidic device. DNA cleavage activity and the aggregation of Cas enzymes was completely avoided under the optimized synthetic conditions. The optimized formulation, which was identified through 2 steps of design of experiments, exhibited excellent gene disruption (up to 97%) and base substitution (up to 23%) without any apparent cytotoxicity. The addition of negative charges to the RNPs by complexing single-stranded oligonucleotide (ssON) significantly enhanced the delivery of both Cas9 and Cpf1 RNPs. The optimized formulation significantly suppressed both HBV DNA and covalently closed circular DNA (cccDNA) in HBV-infected human liver cells compared to adeno-associated virus type 2 (AAV2). These findings represent a significant contribution to the development of CRISPR/Cas RNP delivery technology and its practical applications in genome editing therapy.
Collapse
Affiliation(s)
- Yuichi Suzuki
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | - Haruno Onuma
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | - Risa Sato
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | - Yusuke Sato
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan.
| | - Akari Hashiba
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | - Masatoshi Maeki
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Hokkaido, Japan; JST PRESTO, Saitama, Japan
| | - Manabu Tokeshi
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Hokkaido, Japan; Innovative Research Center for Preventive Medical Engineering, Nagoya University, Nagoya, Japan; Institute of Nano-Life Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya, Japan
| | - Mohammad Enamul Hoque Kayesh
- Laboratory of Animal Hygiene, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan; Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan; Department of Microbiology and Public Health, Patuakhali Science and Technology University, Patuakhali, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Laboratory of Animal Hygiene, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan; Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan.
| |
Collapse
|
42
|
Zhao X, Chen J, Qiu M, Li Y, Glass Z, Xu Q. Imidazole‐Based Synthetic Lipidoids for In Vivo mRNA Delivery into Primary T Lymphocytes. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Xuewei Zhao
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Min Qiu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Yamin Li
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| |
Collapse
|
43
|
Liu Z, Zhou W, Qi C, Kong T. Interface Engineering in Multiphase Systems toward Synthetic Cells and Organelles: From Soft Matter Fundamentals to Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002932. [PMID: 32954548 DOI: 10.1002/adma.202002932] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/19/2020] [Indexed: 06/11/2023]
Abstract
Synthetic cells have a major role in gaining insight into the complex biological processes of living cells; they also give rise to a range of emerging applications from gene delivery to enzymatic nanoreactors. Living cells rely on compartmentalization to orchestrate reaction networks for specialized and coordinated functions. Principally, the compartmentalization has been an essential engineering theme in constructing cell-mimicking systems. Here, efforts to engineer liquid-liquid interfaces of multiphase systems into membrane-bounded and membraneless compartments, which include lipid vesicles, polymer vesicles, colloidosomes, hybrids, and coacervate droplets, are summarized. Examples are provided of how these compartments are designed to imitate biological behaviors or machinery, including molecule trafficking, growth, fusion, energy conversion, intercellular communication, and adaptivity. Subsequently, the state-of-art applications of these cell-inspired synthetic compartments are discussed. Apart from being simplified and cell models for bridging the gap between nonliving matter and cellular life, synthetic compartments also are utilized as intracellular delivery vehicles for nuclei acids and nanoreactors for biochemical synthesis. Finally, key challenges and future directions for achieving the full potential of synthetic cells are highlighted.
Collapse
Affiliation(s)
- Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Wen Zhou
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Cheng Qi
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen, 518000, China
| | - Tiantian Kong
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong, 518000, China
| |
Collapse
|
44
|
Lan X, Zhu W, Huang X, Yu Y, Xiao H, Jin L, Pu JJ, Xie X, She J, Lui VWY, Chen HJ, Su YX. Microneedles loaded with anti-PD-1-cisplatin nanoparticles for synergistic cancer immuno-chemotherapy. NANOSCALE 2020; 12:18885-18898. [PMID: 32902555 DOI: 10.1039/d0nr04213g] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Programmed cell death protein-1 (PD-1) on T-cells combined with programmed cell death ligand-1 (PD-L1) critically accounts for tumor immune evasion. Anti-PD-1 (aPD-1) blocks the binding of PD-1 to PD-L1, thus allowing T-cell activation for tumor cell eradication. Currently, the major challenges for cancer immunotherapy are how to improve the response rate and overcome drug resistance. Dermal administration turns out to be a promising route for immunotherapy since skin is a highly active immune organ containing a large population of resident antigen-presenting cells. Microneedle arrays can pierce the immune-cell-rich epidermis, leading to a robust T-cell response in the microenvironment of tumor cells. Herein, we successfully developed a microneedle patch loaded with pH-responsive tumor-targeted lipid nanoparticles (NPs), which allows local delivery of aPD-1 and cisplatin (CDDP) precisely to cancer tissues for cancer therapy. For in vivo studies, aPD-1/CDDP@NPs delivered through microneedles effectively boosted the immune response, thereby a remarkable effect on tumor regression was realized. Synergistic anticancer mechanisms were therefore activated through robust microneedle-induced T-cell response, blockage of PD-1 in T-cells by aPD-1, and an increase in direct cytotoxicity of CDDP in tumor cells. Strikingly, transdermal delivery using MNs increased the response rate in the animal model unresponsive to aPD-1 systemic therapy. This exhibited promise in the treatment of immunotherapy-unresponsive cancers. Taken together, microneedle-mediated local delivery of nano-encapsulated chemotherapeutic and immunotherapeutic agents at tumor skin sites provides a novel treatment strategy and insights into cancer therapy.
Collapse
Affiliation(s)
- Xinmiao Lan
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
He W, Xing X, Wang X, Wu D, Wu W, Guo J, Mitragotri S. Nanocarrier‐Mediated Cytosolic Delivery of Biopharmaceuticals. ADVANCED FUNCTIONAL MATERIALS 2020; 30. [DOI: 10.1002/adfm.201910566] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/16/2020] [Indexed: 01/04/2025]
Abstract
AbstractBiopharmaceuticals have emerged to play a vital role in disease treatment and have shown promise in the rapidly expanding pharmaceutical market due to their high specificity and potency. However, the delivery of these biologics is hindered by various physiological barriers, owing primarily to the poor cell membrane permeability, low stability, and increased size of biologic agents. Since many biological drugs are intended to function by interacting with intracellular targets, their delivery to intracellular targets is of high relevance. In this review, the authors summarize and discuss the use of nanocarriers for intracellular delivery of biopharmaceuticals via endosomal escape and, especially, the routes of direct cytosolic delivery by means including the caveolae‐mediated pathway, contact release, intermembrane transfer, membrane fusion, direct translocation, and membrane disruption. Strategies with high potential for translation are highlighted. Finally, the authors conclude with the clinical translation of promising carriers and future perspectives.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xuyang Xing
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xiaoling Wang
- School of Biomass Science and Engineering Sichuan University Chengdu 610065 China
| | - Debra Wu
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of Ministry of Education of China School of Pharmacy Fudan University Shanghai 201203 China
| | - Junling Guo
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| |
Collapse
|
46
|
Zhao X, Chen J, Qiu M, Li Y, Glass Z, Xu Q. Imidazole‐Based Synthetic Lipidoids for In Vivo mRNA Delivery into Primary T Lymphocytes. Angew Chem Int Ed Engl 2020; 59:20083-20089. [DOI: 10.1002/anie.202008082] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Xuewei Zhao
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Min Qiu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Yamin Li
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| |
Collapse
|
47
|
Li Y, Jarvis R, Zhu K, Glass Z, Ogurlu R, Gao P, Li P, Chen J, Yu Y, Yang Y, Xu Q. Protein and mRNA Delivery Enabled by Cholesteryl-Based Biodegradable Lipidoid Nanoparticles. Angew Chem Int Ed Engl 2020; 59:14957-14964. [PMID: 32438474 PMCID: PMC7679290 DOI: 10.1002/anie.202004994] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Indexed: 11/12/2022]
Abstract
Developing safe and efficient delivery systems for therapeutic biomacromolecules is a long-standing challenge. Herein, we report a newly developed combinatorial library of cholesteryl-based disulfide bond-containing biodegradable cationic lipidoid nanoparticles. We have identified a subset of this library which is effective for protein and mRNA delivery in vitro and in vivo. These lipidoids showed comparable transfection efficacies but much lower cytotoxicities compared to the Lpf2k in vitro. In vivo studies in adult mice demonstrated the successful delivery of genome engineering protein and mRNA molecules in the skeletal muscle (via intramuscular injection), lung and spleen (via intravenous injection), and brain (via lateral ventricle infusion).
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Rachel Jarvis
- Department of Neuroscience, Tufts University, Boston, MA 02111, USA
| | - Kuixin Zhu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Zachary Glass
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Roza Ogurlu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Peiyang Gao
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Peixuan Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yingjie Yu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yongjie Yang
- Department of Neuroscience, Tufts University, Boston, MA 02111, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
48
|
Li Y, Li R, Chakraborty A, Ogurlu R, Zhao X, Chen J, Xu Q. Combinatorial Library of Cyclic Benzylidene Acetal-Containing pH-Responsive Lipidoid Nanoparticles for Intracellular mRNA Delivery. Bioconjug Chem 2020; 31:1835-1843. [PMID: 32520527 DOI: 10.1021/acs.bioconjchem.0c00295] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lipidoid nanoparticles have been demonstrated to be effective for intracellular delivery of small molecule drugs, proteins, and nucleic acids. Stimuli-responsive lipidoid nanoparticles are able to further improve delivery efficacy and reduce carrier-induced toxicity. Our group previously developed reduction and photoresponsive combinatorial libraries of lipidoid nanoparticles for small molecule and biologics delivery. Herein, we describe the synthesis, characterization, and intracellular mRNA delivery application of a new library of pH-responsive lipidoid nanoparticles. The acid-degradable cyclic benzylidene acetal-containing cationic lipidoids (R-O16CBA) were synthesized through a multistep reaction and characterized by NMR and MS. The acid-triggered degradation of lipidoids was studied using NMR, MS, DLS, and TEM. The results revealed that the R-O16CBA lipidoid can be completely degraded at pH 5. The R-O16CBA lipidoid nanoparticles were then fabricated with different formulations of DOPE and cholesterol and tested in vitro for intracellular mRNA delivery.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Raissa Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Anirban Chakraborty
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Roza Ogurlu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Xuewei Zhao
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
49
|
Li Y, Jarvis R, Zhu K, Glass Z, Ogurlu R, Gao P, Li P, Chen J, Yu Y, Yang Y, Xu Q. Protein and mRNA Delivery Enabled by Cholesteryl‐Based Biodegradable Lipidoid Nanoparticles. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Rachel Jarvis
- Department of Neuroscience Tufts University Boston MA 02111 USA
| | - Kuixin Zhu
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Roza Ogurlu
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Peiyang Gao
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Peixuan Li
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Yingjie Yu
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| | - Yongjie Yang
- Department of Neuroscience Tufts University Boston MA 02111 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| |
Collapse
|
50
|
|