1
|
Chen B, Ren Q, Jiang P, Wu Q, Shuai Q, Yan Y. Combinatorial Synthesis of Alkyl Chain-Capped Poly(β-Amino Ester)s for Effective siRNA Delivery. Macromol Biosci 2024; 24:e2400168. [PMID: 39052313 DOI: 10.1002/mabi.202400168] [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: 04/08/2024] [Revised: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Poly (β-amino ester) (PBAE) is a class of biodegradable polymers containing ester bonds in their main chain, extensively investigated as cationic polymer carriers for siRNA. Most current PBAE carriers rely on termination with hydrophilic or charged amines. In this study, a polymer platform consisting of 168 PBAE polymers with hydrophobic alkyl chain terminals is constructed through sequential aza-Michael addition. A large number of effective carriers are identified through in vitro screening of the PBAE platform for siLuc delivery to HeLa-Luc cells. Specifically, PA8-C6 and PA8-C8 achieve remarkable gene knockdown efficacies of up to 80% with low cytotoxicity. Certain materials from the PA2 and PA5 series demonstrate potent siRNA delivery capabilities associated with elevated cytotoxicity. The pKa value of PBAE is predominantly determined by the hydrophilic amine side chains rather than the end-capping groups. A pKa range of ≈6.2-6.5 may contribute to the excellent delivery capability for PA8 series carriers. The co-formulation of PBAE carriers with helper lipids leads to the reduced size and surface charges of the polyplex NPs with siRNA, consequently decreasing the cytotoxicity and enhancing siRNA delivery efficacy. These findings hold significant implications for the development of novel degradable polymer carriers for siRNA delivery.
Collapse
Affiliation(s)
- Baiqiu Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qidi Ren
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Pingge Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qiong Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qi Shuai
- College of Pharmaceutical Sciences and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
2
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
3
|
Cheng Q, Farbiak L, Vaidya A, Guerrero E, Lee EE, Rose EK, Wang X, Robinson J, Lee SM, Wei T, Miller WE, Alvarez Benedicto E, Lian X, Wang RC, Siegwart DJ. In situ production and secretion of proteins endow therapeutic benefit against psoriasiform dermatitis and melanoma. Proc Natl Acad Sci U S A 2023; 120:e2313009120. [PMID: 38109533 PMCID: PMC10756300 DOI: 10.1073/pnas.2313009120] [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: 07/28/2023] [Accepted: 10/30/2023] [Indexed: 12/20/2023] Open
Abstract
Genetic medicines have the potential to treat various diseases; however, certain ailments including inflammatory diseases and cancer would benefit from control over extracellular localization of therapeutic proteins. A critical gap therefore remains the need to develop and incorporate methodologies that allow for posttranslational control over expression dynamics, localization, and stability of nucleic acid-generated protein therapeutics. To address this, we explored how the body's endogenous machinery controls protein localization through signal peptides (SPs), including how these motifs could be incorporated modularly into therapeutics. SPs serve as a virtual zip code for mRNA transcripts that direct the cell where to send completed proteins within the cell and the body. Utilizing this signaling biology, we incorporated secretory SP sequences upstream of mRNA transcripts coding for reporter, natural, and therapeutic proteins to induce secretion of the proteins into systemic circulation. SP sequences generated secretion of various engineered proteins into the bloodstream following intravenous, intramuscular, and subcutaneous SP mRNA delivery by lipid, polymer, and ionizable phospholipid delivery carriers. SP-engineered etanercept/TNF-α inhibitor proteins demonstrated therapeutic efficacy in an imiquimod-induced psoriasis model by reducing hyperkeratosis and inflammation. An SP-engineered anti-PD-L1 construct mediated mRNA encoded proteins with longer serum half-lives that reduced tumor burden and extended survival in MC38 and B16F10 cancer models. The modular nature of SP platform should enable intracellular and extracellular localization control of various functional proteins for diverse therapeutic applications.
Collapse
Affiliation(s)
- Qiang Cheng
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Lukas Farbiak
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Amogh Vaidya
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Erick Guerrero
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Eunice E. Lee
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Elysha K. Rose
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Xu Wang
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Joshua Robinson
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Sang M. Lee
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Tuo Wei
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - William E. Miller
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Ester Alvarez Benedicto
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Xizhen Lian
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Richard C. Wang
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Daniel J. Siegwart
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX75390
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
4
|
Dilliard SA, Siegwart DJ. Passive, active and endogenous organ-targeted lipid and polymer nanoparticles for delivery of genetic drugs. NATURE REVIEWS. MATERIALS 2023; 8:282-300. [PMID: 36691401 PMCID: PMC9850348 DOI: 10.1038/s41578-022-00529-7] [Citation(s) in RCA: 237] [Impact Index Per Article: 118.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 05/03/2023]
Abstract
Genetic drugs based on nucleic acid biomolecules are a rapidly emerging class of medicines that directly reprogramme the central dogma of biology to prevent and treat disease. However, multiple biological barriers normally impede the intracellular delivery of nucleic acids, necessitating the use of a delivery system. Lipid and polymer nanoparticles represent leading approaches for the clinical translation of genetic drugs. These systems circumnavigate biological barriers and facilitate the intracellular delivery of nucleic acids in the correct cells of the target organ using passive, active and endogenous targeting mechanisms. In this Review, we highlight the constituent materials of these advanced nanoparticles, their nucleic acid cargoes and how they journey through the body. We discuss targeting principles for liver delivery, as it is the organ most successfully targeted by intravenously administered nanoparticles to date, followed by the expansion of these concepts to extrahepatic (non-liver) delivery. Ultimately, this Review connects emerging materials and biological insights playing key roles in targeting specific organs and cells in vivo.
Collapse
Affiliation(s)
- Sean A. Dilliard
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX USA
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX USA
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Daniel J. Siegwart
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX USA
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX USA
- Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX USA
| |
Collapse
|
5
|
Yan Y, Zhu F, Su H, Liu X, Ren Q, Huang F, Ye W, Zhao M, Zhao Y, Zhao J, Shuai Q. Construction of Degradable and Amphiphilic Triblock Polymer Carriers for Effective Delivery of siRNA. Macromol Biosci 2022; 22:e2200232. [PMID: 36086889 DOI: 10.1002/mabi.202200232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/17/2022] [Indexed: 01/15/2023]
Abstract
The development of effective and safe delivery carriers is one of the prerequisites for the clinical translation of siRNA-based therapeutics. In this study, a library of 144 functional triblock polymers using ring-opening polymerization (ROP) and thiol-ene click reaction is constructed. These triblock polymers are composed of hydrophilic poly (ethylene oxide) (PEO), hydrophobic poly (ε-caprolactone) (PCL), and cationic amine blocks. Three effective carriers are discovered by high-throughput screening of these polymers for siRNA delivery to HeLa-Luc cells. In vitro evaluation shows that siLuc-loaded nanoparticles (NPs) fabricated with leading polymer carriers exhibit sufficient knockdown of luciferase genes and relatively low cytotoxicity. The chemical structure of polymers significantly affects the physicochemical properties of the resulting siRNA-loaded NPs, which leads to different cellular uptake of NPs and endosomal escape of loaded siRNA and thus the overall in vitro siRNA delivery efficacy. After systemic administration to mice with xenograft tumors, siRNA NPs based on P2-4.5A8 are substantially accumulated at tumor sites, suggesting that PEO and PCL blocks are beneficial for improving blood circulation and biodistribution of siRNA NPs. This functional triblock polymer platform may have great potential in the development of siRNA-based therapies for the treatment of cancers.
Collapse
Affiliation(s)
- Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| | - Fangtao Zhu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| | - Huahui Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| | - Xiaomin Liu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| | - Qidi Ren
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| | - Fangqian Huang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| | - Wenbo Ye
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| | - Mengdan Zhao
- Women's Hospital, School of Medicine, Zhejiang University and Key Laboratory of Women's Reproductive Health Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, P. R. China
| | - Yunchun Zhao
- Women's Hospital, School of Medicine, Zhejiang University and Key Laboratory of Women's Reproductive Health Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, P. R. China
| | - Junpeng Zhao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, Guangdong, 510641, P. R. China
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China
| |
Collapse
|
6
|
Mashima R, Takada S. Lipid Nanoparticles: A Novel Gene Delivery Technique for Clinical Application. Curr Issues Mol Biol 2022; 44:5013-5027. [PMID: 36286056 PMCID: PMC9600891 DOI: 10.3390/cimb44100341] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/08/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Lipid nanoparticles (LNPs) are an emerging vehicle for gene delivery that accommodate both nucleic acid and protein. Based on the experience of therapeutic liposomes, current LNPs have been developed based on the chemistry of lipids and RNA and on the biology of human disease. LNPs have been used for the development of Onpattro, an siRNA drug for transthyretin-mediated amyloidosis, in 2018. The subsequent outbreak of COVID-19 required a vaccine for its suppression. LNP-based vaccine production received much attention for this and resulted in great success. In this review, the essential technology of LNP gene delivery has been described according to the chemistry for LNP production and biology for its clinical application.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
- Correspondence:
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| |
Collapse
|
7
|
Álvarez-Benedicto E, Farbiak L, Márquez Ramírez M, Wang X, Johnson LT, Mian O, Guerrero ED, Siegwart DJ. Optimization of phospholipid chemistry for improved lipid nanoparticle (LNP) delivery of messenger RNA (mRNA). Biomater Sci 2022; 10:549-559. [PMID: 34904974 PMCID: PMC9113778 DOI: 10.1039/d1bm01454d] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lipid nanoparticles (LNPs) have been established as an essential platform for nucleic acid delivery. Efforts have led to the development of vaccines that protect against SARS-CoV-2 infection using LNPs to deliver messenger RNA (mRNA) coding for the viral spike protein. Out of the four essential components that comprise LNPs, phospholipids represent an underappreciated opportunity for fundamental and translational study. We investigated this avenue by systematically modulating the identity of the phospholipid in LNPs with the goal of identifying specific moieties that directly enhance or hinder delivery efficacy. Results indicate that phospholipid chemistry can enhance mRNA delivery by increasing membrane fusion and enhancing endosomal escape. Phospholipids containing phosphoethanolamine (PE) head groups likely increase endosomal escape due to their fusogenic properties. Additionally, it was found that zwitterionic phospholipids mainly aided liver delivery, whereas negatively charged phospholipids changed the tropism of the LNPs from liver to spleen. These results demonstrate that the choice of phospholipid plays a role intracellularly by enhancing endosomal escape, while also driving organ tropism in vivo. These findings were then applied to Selective Organ Targeting (SORT) LNPs to manipulate and control spleen-specific delivery. Overall, selection of the phospholipid in LNPs provides an important handle to design and optimize LNPs for improved mRNA delivery and more effective therapeutics.
Collapse
Affiliation(s)
- Ester Álvarez-Benedicto
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Lukas Farbiak
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Martha Márquez Ramírez
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Xu Wang
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Lindsay T Johnson
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Osamah Mian
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Erick D Guerrero
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | - Daniel J Siegwart
- Department of Biochemistry, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| |
Collapse
|
8
|
Chen M, Wang H, Guo H, Zhang Y, Chen L. Systematic Investigation of Biocompatible Cationic Polymeric Nucleic Acid Carriers for Immunotherapy of Hepatocellular Carcinoma. Cancers (Basel) 2021; 14:85. [PMID: 35008249 PMCID: PMC8750096 DOI: 10.3390/cancers14010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third-largest cause of cancer death worldwide, while immunotherapy is rapidly being developed to fight HCC with great potential. Nucleic acid drugs are the most important modulators in HCC immunotherapy. To boost the efficacy of therapeutics and amplify the efficiency of genetic materials, biocompatible polymers are commonly used. However, under the strong need of a summary for current developments of biocompatible polymeric nucleic acid carriers for immunotherapy of HCC, there is rare review article specific to this topic to our best knowledge. In this article, we will discuss the current progress of immunotherapy for HCC, biocompatible cationic polymers (BCPs) as nucleic acid carriers used (or potential) to fight HCC, the roles of biocompatible polymeric carriers for nucleic acid delivery, and nucleic acid delivery by biocompatible polymers for immunotherapy. At the end, we will conclude the review and discuss future perspectives. This article discusses biocompatible polymeric nucleic acid carriers for immunotherapy of HCC from multidiscipline perspectives and provides a new insight in this domain. We believe this review will be interesting to polymer chemists, pharmacists, clinic doctors, and PhD students in related disciplines.
Collapse
Affiliation(s)
- Mingsheng Chen
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| | - Hao Wang
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| | - Hongying Guo
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| | - Ying Zhang
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Liang Chen
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| |
Collapse
|
9
|
Yu X, Liu S, Cheng Q, Lee SM, Wei T, Zhang D, Farbiak L, Johnson LT, Wang X, Siegwart DJ. Hydrophobic Optimization of Functional Poly(TPAE-co-suberoyl chloride) for Extrahepatic mRNA Delivery following Intravenous Administration. Pharmaceutics 2021; 13:pharmaceutics13111914. [PMID: 34834329 PMCID: PMC8624493 DOI: 10.3390/pharmaceutics13111914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023] Open
Abstract
Messenger RNA (mRNA) has generated great attention due to its broad potential therapeutic applications, including vaccines, protein replacement therapy, and immunotherapy. Compared to other nucleic acids (e.g., siRNA and pDNA), there are more opportunities to improve the delivery efficacy of mRNA through systematic optimization. In this report, we studied a high-throughput library of 1200 functional polyesters for systemic mRNA delivery. We focused on the chemical investigation of hydrophobic optimization as a method to adjust mRNA polyplex stability, diameter, pKa, and efficacy. Focusing on a region of the library heatmap (PE4K-A17), we further explored the delivery of luciferase mRNA to IGROV1 ovarian cancer cells in vitro and to C57BL/6 mice in vivo following intravenous administration. PE4K-A17-0.2C8 was identified as an efficacious carrier for delivering mRNA to mouse lungs. The delivery selectivity between organs (lungs versus spleen) was found to be tunable through chemical modification of polyesters (both alkyl chain length and molar ratio in the formulation). Cre recombinase mRNA was delivered to the Lox-stop-lox tdTomato mouse model to study potential application in gene editing. Overall, we identified a series of polymer-mRNA polyplexes stabilized with Pluronic F-127 for safe and effective delivery to mouse lungs and spleens. Structure–activity relationships between alkyl side chains and in vivo delivery were elucidated, which may be informative for the continued development of polymer-based mRNA delivery.
Collapse
|
10
|
Abstract
RNA-based therapeutics have shown great promise in treating a broad spectrum of diseases through various mechanisms including knockdown of pathological genes, expression of therapeutic proteins, and programmed gene editing. Due to the inherent instability and negative-charges of RNA molecules, RNA-based therapeutics can make the most use of delivery systems to overcome biological barriers and to release the RNA payload into the cytosol. Among different types of delivery systems, lipid-based RNA delivery systems, particularly lipid nanoparticles (LNPs), have been extensively studied due to their unique properties, such as simple chemical synthesis of lipid components, scalable manufacturing processes of LNPs, and wide packaging capability. LNPs represent the most widely used delivery systems for RNA-based therapeutics, as evidenced by the clinical approvals of three LNP-RNA formulations, patisiran, BNT162b2, and mRNA-1273. This review covers recent advances of lipids, lipid derivatives, and lipid-derived macromolecules used in RNA delivery over the past several decades. We focus mainly on their chemical structures, synthetic routes, characterization, formulation methods, and structure-activity relationships. We also briefly describe the current status of representative preclinical studies and clinical trials and highlight future opportunities and challenges.
Collapse
Affiliation(s)
- Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Changzhen Sun
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chang Wang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Katarina E Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
11
|
Rinoldi C, Zargarian SS, Nakielski P, Li X, Liguori A, Petronella F, Presutti D, Wang Q, Costantini M, De Sio L, Gualandi C, Ding B, Pierini F. Nanotechnology-Assisted RNA Delivery: From Nucleic Acid Therapeutics to COVID-19 Vaccines. SMALL METHODS 2021; 5:e2100402. [PMID: 34514087 PMCID: PMC8420172 DOI: 10.1002/smtd.202100402] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/04/2021] [Indexed: 05/07/2023]
Abstract
In recent years, the main quest of science has been the pioneering of the groundbreaking biomedical strategies needed for achieving a personalized medicine. Ribonucleic acids (RNAs) are outstanding bioactive macromolecules identified as pivotal actors in regulating a wide range of biochemical pathways. The ability to intimately control the cell fate and tissue activities makes RNA-based drugs the most fascinating family of bioactive agents. However, achieving a widespread application of RNA therapeutics in humans is still a challenging feat, due to both the instability of naked RNA and the presence of biological barriers aimed at hindering the entrance of RNA into cells. Recently, material scientists' enormous efforts have led to the development of various classes of nanostructured carriers customized to overcome these limitations. This work systematically reviews the current advances in developing the next generation of drugs based on nanotechnology-assisted RNA delivery. The features of the most used RNA molecules are presented, together with the development strategies and properties of nanostructured vehicles. Also provided is an in-depth overview of various therapeutic applications of the presented systems, including coronavirus disease vaccines and the newest trends in the field. Lastly, emerging challenges and future perspectives for nanotechnology-mediated RNA therapies are discussed.
Collapse
Affiliation(s)
- Chiara Rinoldi
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Pawel Nakielski
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Xiaoran Li
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Anna Liguori
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
| | - Francesca Petronella
- Institute of Crystallography CNR‐ICNational Research Council of ItalyVia Salaria Km 29.300Monterotondo – Rome00015Italy
| | - Dario Presutti
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Qiusheng Wang
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Marco Costantini
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Luciano De Sio
- Department of Medico‐Surgical Sciences and BiotechnologiesResearch Center for BiophotonicsSapienza University of RomeCorso della Repubblica 79Latina04100Italy
- CNR‐Lab. LicrylInstitute NANOTECArcavacata di Rende87036Italy
| | - Chiara Gualandi
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
- Interdepartmental Center for Industrial Research on Advanced Applications in Mechanical Engineering and Materials TechnologyCIRI‐MAMUniversity of BolognaViale Risorgimento 2Bologna40136Italy
| | - Bin Ding
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Filippo Pierini
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| |
Collapse
|
12
|
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
|
13
|
Abd Elwakil MM, Gao T, Isono T, Sato Y, Elewa YHA, Satoh T, Harashima H. Engineered ε-decalactone lipomers bypass the liver to selectively in vivo deliver mRNA to the lungs without targeting ligands. MATERIALS HORIZONS 2021; 8:2251-2259. [PMID: 34846429 DOI: 10.1039/d1mh00185j] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
RNA drugs hold real potential for tackling devastating diseases that are currently resistant to small molecule drugs or monoclonal antibodies. However, since these drugs are unstable in vivo and unable to pass through cellular membranes their clinical realization is limited by their successful delivery to target sites. Herein we report on the design of a combinatorial library of polyester lipomers based on the renewable monomer, ε-decalactone (ε-DL), via organocatalytic ring-opening polymerization for mRNA delivery. The ε-DL lipomers showed a surprisingly efficient ability to target the lungs upon intravenous administration. Interestingly, most of the lipomers achieved functional EGFP expression in the lungs, while minimally transfecting hepatocytes and splenic cells. This simple approach for the design of biodegradable materials has the potential for the clinical translation of genetic medicines for the treatment of lung diseases.
Collapse
Affiliation(s)
- Mahmoud M Abd Elwakil
- Laboratory of innovative nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Hokkaido 060-0812, Japan.
| | | | | | | | | | | | | |
Collapse
|
14
|
Dobrowolski C, Paunovska K, Hatit MZC, Lokugamage MP, Dahlman JE. Therapeutic RNA Delivery for COVID and Other Diseases. Adv Healthc Mater 2021; 10:e2002022. [PMID: 33661555 PMCID: PMC7995096 DOI: 10.1002/adhm.202002022] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/11/2021] [Indexed: 12/11/2022]
Abstract
RNA can alter the expression of endogenous genes and can be used to express therapeutic proteins. As a result, RNA-based therapies have recently mitigated disease in patients. Yet most potential RNA therapies cannot currently be developed, in large part because delivering therapeutic quantities of RNA drugs to diseased cells remains difficult. Here, recent studies focused on the biological hurdles that make in vivo drug delivery challenging are described. Then RNA drugs that have overcome these challenges in humans, focusing on siRNA to treat liver disease and mRNA to vaccinate against COVID, are discussed. Finally, research centered on improving drug delivery to new tissues is highlighted, including the development of high-throughput in vivo nanoparticle DNA barcoding assays capable of testing over 100 distinct nanoparticles in a single animal.
Collapse
Affiliation(s)
- Curtis Dobrowolski
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University School of MedicineAtlantaGA30332USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University School of MedicineAtlantaGA30332USA
| | - Marine Z. C. Hatit
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University School of MedicineAtlantaGA30332USA
| | - Melissa P. Lokugamage
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University School of MedicineAtlantaGA30332USA
| | - James E. Dahlman
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University School of MedicineAtlantaGA30332USA
| |
Collapse
|
15
|
Wang Z, Liu Z, Mei J, Xu S, Liu Y. The next generation therapy for lung cancer: taking medicine by inhalation. NANOTECHNOLOGY 2021; 32:392002. [PMID: 34167099 DOI: 10.1088/1361-6528/ac0e68] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
The inhalation administration method which has been applied to treat respiratory diseases has the characteristics of painlessness high efficiency and non-invasiveness, and the drug can also be targeted at the organ level first to reduce the loss of drug during circulation. Therefore, delivering medicine by inhalation administration has brought a new turnaround for lung cancer treatment. Herein from the perspective of combining traditional drug delivery design strategies with new drug delivery methods how to improve lung targeting efficiency and treatment efficacy is discussed. We also discuss the comparative advantages of inhaled drug delivery and traditional administration in the treatment of lung cancer such as intravenous injection. And the researches are divided into different forms of inhalation administration studied in the treatment of lung cancer in recent years, such as single-component loaded and multi-component loaded systems and their advantages. Finally, the obstacles of the application of carrier materials for inhalation administration and the prospects for improvement of lung cancer treatment methods are presented.
Collapse
Affiliation(s)
- Ziyao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zifan Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Jie Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- GBA National Institute for Nanotechnology Innovation, Guangdong 510700, People's Republic of China
| |
Collapse
|
16
|
Farbiak L, Cheng Q, Wei T, Álvarez-Benedicto E, Johnson LT, Lee S, Siegwart DJ. All-In-One Dendrimer-Based Lipid Nanoparticles Enable Precise HDR-Mediated Gene Editing In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006619. [PMID: 34137093 PMCID: PMC10041668 DOI: 10.1002/adma.202006619] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/19/2021] [Indexed: 05/08/2023]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) protein gene editing is poised to transform the treatment of genetic diseases. However, limited progress has been made toward precise editing of DNA via homology-directed repair (HDR) that requires careful orchestration of complex steps. Herein, dendrimer-based lipid nanoparticles (dLNPs) are engineered to co-encapsulate and deliver multiple components for in vivo HDR correction. BFP/GFP switchable HEK293 cells with a single Y66H amino acid mutation are employed to assess HDR-mediated gene editing following simultaneous, one-pot delivery of Cas9 mRNA, single-guide RNA, and donor DNA. Molar ratios of individual LNP components and weight ratios of the three nucleic acids are systematically optimized to increase HDR efficiency. Using flow cytometry, fluorescence imaging, and DNA sequencing to quantify editing, optimized 4A3-SC8 dLNPs edit >91% of all cells with 56% HDR efficiency in vitro and >20% HDR efficiency in xenograft tumors in vivo. Due to the all-in-one simplicity and high efficacy, the developed dLNPs offer a promising route toward the gene correction of disease-causing mutations.
Collapse
Affiliation(s)
- Lukas Farbiak
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qiang Cheng
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tuo Wei
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ester Álvarez-Benedicto
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lindsay T Johnson
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sang Lee
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Siegwart
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
17
|
Rawal S, Patel M. Bio-Nanocarriers for Lung Cancer Management: Befriending the Barriers. NANO-MICRO LETTERS 2021; 13:142. [PMID: 34138386 PMCID: PMC8196938 DOI: 10.1007/s40820-021-00630-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/23/2021] [Indexed: 05/03/2023]
Abstract
Lung cancer is a complex thoracic malignancy developing consequential to aberrations in a myriad of molecular and biomolecular signaling pathways. It is one of the most lethal forms of cancers accounting to almost 1.8 million new annual incidences, bearing overall mortality to incidence ratio of 0.87. The dismal prognostic scenario at advanced stages of the disease and metastatic/resistant tumor cell populations stresses the requisite of advanced translational interdisciplinary interventions such as bionanotechnology. This review article deliberates insights and apprehensions on the recent prologue of nanobioengineering and bionanotechnology as an approach for the clinical management of lung cancer. The role of nanobioengineered (bio-nano) tools like bio-nanocarriers and nanobiodevices in secondary prophylaxis, diagnosis, therapeutics, and theranostics for lung cancer management has been discussed. Bioengineered, bioinspired, and biomimetic bio-nanotools of considerate translational value have been reviewed. Perspectives on existent oncostrategies, their critical comparison with bio-nanocarriers, and issues hampering their clinical bench side to bed transformation have also been summarized.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India
| | - Mayur Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
18
|
Alsudir SA, Almalik A, Alhasan AH. Catalogue of self-targeting nano-medical inventions to accelerate clinical trials. Biomater Sci 2021; 9:3898-3910. [PMID: 33912874 DOI: 10.1039/d1bm00235j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Repetitive outbreaks and prolonged epidemics represent mortal threats to global health, creating chaos in our globalized world. To date, scientists have been compelled to follow FDA guidelines for conventional clinical trials, which decelerates the release of effective therapies to battle outbreaks and safeguard global health security. Developing multi-purpose platform nanotechnologies to self-target specific organs in response to the disease microenvironment could greatly help to rapidly anticipate and efficiently manage outbreaks. Nano-interventions in the form of self-targeting nanoparticles (NPs) could accelerate the clinical translation of potential drugs to fight future outbreaks via innovating their clinical trials. This review sets the foundation of the self-targeting concept to govern the in vivo fate of NPs without the need to complicate the engineering designs with targeting ligands. The proposed catalogue of accelerated nano-innovations offers self-targeting, physiological trafficking, bio-compliance, and controllable drug release in response to associated smart linkers.
Collapse
Affiliation(s)
- Samar A Alsudir
- National Center for Pharmaceutical Technology, Life science and Environmental Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11461, Saudi Arabia.
| | - Abdulaziz Almalik
- National Center for Pharmaceutical Technology, Life science and Environmental Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11461, Saudi Arabia. and KACST-BWH/Harvard Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11461, Saudi Arabia
| | - Ali H Alhasan
- National Center for Pharmaceutical Technology, Life science and Environmental Research Institute, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11461, Saudi Arabia. and KACST-BWH/Harvard Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11461, Saudi Arabia and College of Science and General Studies, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| |
Collapse
|
19
|
Responsive Polyesters with Alkene and Carboxylic Acid Side-Groups for Tissue Engineering Applications. Polymers (Basel) 2021; 13:polym13101636. [PMID: 34070123 PMCID: PMC8158382 DOI: 10.3390/polym13101636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 11/17/2022] Open
Abstract
Main chain polyesters have been extensively used in the biomedical field. Despite their many advantages, including biocompatibility, biodegradability, and others, these materials are rather inert and lack specific functionalities which will endow them with additional biological and responsive properties. In this work, novel pH-responsive main chain polyesters have been prepared by a conventional condensation polymerization of a vinyl functionalized diol with a diacid chloride, followed by a photo-induced thiol-ene click reaction to attach functional carboxylic acid side-groups along the polymer chains. Two different mercaptocarboxylic acids were employed, allowing to vary the alkyl chain length of the polymer pendant groups. Moreover, the degree of modification, and as a result, the carboxylic acid content of the polymers, was easily tuned by varying the irradiation time during the click reaction. Both these parameters, were shown to strongly influence the responsive behavior of the polyesters, which presented adjustable pKα values and water solubilities. Finally, the difunctional polyesters bearing the alkene and carboxylic acid functionalities enabled the preparation of cross-linked polyester films by chemically linking the pendant vinyl bonds on the polymer side groups. The biocompatibility of the cross-linked polymers films was assessed in L929 fibroblast cultures and showed that the cell viability, proliferation, and attachment were greatly promoted on the polyester surface, bearing the shorter alkyl chain length side groups and the higher fraction of carboxylic acid functionalities.
Collapse
|
20
|
de Braganca L, Ferguson GJ, Luis Santos J, Derrick JP. Adverse immunological responses against non-viral nanoparticle (NP) delivery systems in the lung. J Immunotoxicol 2021; 18:61-73. [PMID: 33956565 PMCID: PMC8788408 DOI: 10.1080/1547691x.2021.1902432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
There is a large, unmet medical need to treat chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis and other respiratory diseases. New modalities are being developed, including gene therapy which treats the disease at the DNA/RNA level. Despite recent innovations in non-viral gene therapy delivery for chronic respiratory diseases, unwanted or adverse interactions with immune cells, particularly macrophages, can limit drug efficacy. This review will examine the relationship between the design and fabrication of non-viral nucleic acid nanoparticle (NP) delivery systems and their ability to trigger unwanted immunogenic responses in lung tissues. NP formulated with peptides, lipids, synthetic and natural polymers provide a robust means of delivering the genetic cargos to the desired cells. However NP, or their components, may trigger local responses such as cell damage, edema, inflammation, and complement activation. These effects may be acute short-term reactions or chronic long-term effects like fibrosis, increased susceptibility to diseases, autoimmune disorders, and even cancer. This review examines the relationship between physicochemical properties, i.e. shape, charge, hydrophobicity, composition and stiffness, and interactions of NP with pulmonary immune cells. Inhalation is the ideal route of administration for direct delivery but inhaled NP encounter innate immune cells, such as alveolar macrophages (AM) and dendritic cells (DC), that perceive them as harmful foreign material, interfere with gene delivery to target cells, and can induce undesirable side effects. Recommendations for fabrication and formulation of gene therapies to avoid adverse immunological responses are given. These include fine tuning physicochemical properties, functionalization of the surface of NP to actively target diseased pulmonary cells and employing biomimetics to increase immunotolerance.
Collapse
Affiliation(s)
- Leonor de Braganca
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - G John Ferguson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jose Luis Santos
- Dosage Form Design Development, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Jeremy P Derrick
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Wang H, Zhang S, Lv J, Cheng Y. Design of polymers for siRNA delivery: Recent progress and challenges. VIEW 2021. [DOI: 10.1002/viw.20200026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology School of Molecular Science and Engineering South China University of Technology Guangzhou China
| | - Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology School of Molecular Science and Engineering South China University of Technology Guangzhou China
| | - Jia Lv
- South China Advanced Institute for Soft Matter Science and Technology School of Molecular Science and Engineering South China University of Technology Guangzhou 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 China
- Shanghai Key Laboratory of Regulatory Biology School of Life Sciences East China Normal University Shanghai China
| |
Collapse
|
22
|
Sato Y, Nakamura T, Yamada Y, Harashima H. The nanomedicine rush: New strategies for unmet medical needs based on innovative nano DDS. J Control Release 2021; 330:305-316. [DOI: 10.1016/j.jconrel.2020.12.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
|
23
|
Jiang X, Abedi K, Shi J. Polymeric nanoparticles for RNA delivery. REFERENCE MODULE IN MATERIALS SCIENCE AND MATERIALS ENGINEERING 2021. [PMCID: PMC8568333 DOI: 10.1016/b978-0-12-822425-0.00017-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
As exemplified by recent clinical approval of RNA drugs including the latest COVID-19 mRNA vaccines, RNA therapy has demonstrated great promise as an emerging medicine. Central to the success of RNA therapy is the delivery of RNA molecules into the right cells at the right location. While the clinical success of nanotechnology in RNA therapy has been limited to lipid-based nanoparticles currently, polymers, due to their tunability and robustness, have also evolved as a class of promising material for the delivery of various therapeutics including RNAs. This article overviews different types of polymers used in RNA delivery and the methods for the formulation of polymeric nanoparticles and highlights recent progress of polymeric nanoparticle-based RNA therapy.
Collapse
|
24
|
Chow MYT, Qiu Y, Lam JKW. Inhaled RNA Therapy: From Promise to Reality. Trends Pharmacol Sci 2020; 41:715-729. [PMID: 32893004 PMCID: PMC7471058 DOI: 10.1016/j.tips.2020.08.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
RNA-based medicine is receiving growing attention for its diverse roles and potential therapeutic capacity. The largest obstacle in its clinical translation remains identifying a safe and effective delivery system. Studies investigating RNA therapeutics in pulmonary diseases have rapidly expanded and drug administration by inhalation allows the direct delivery of RNA therapeutics to the target site of action while minimizing systemic exposure. In this review, we highlight recent developments in pulmonary RNA delivery systems with the use of nonviral vectors. We also discuss the major knowledge gaps that require thorough investigation and provide insights that will help advance this exciting field towards the bedside.
Collapse
Affiliation(s)
- Michael Y T Chow
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR; Advanced Drug Delivery Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Yingshan Qiu
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Jenny K W Lam
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
25
|
Abstract
Polyethyleneimine (PEI) has been extensively investigated as an efficient carrier for nucleic acid delivery. Yet, it suffers from a high toxicity profile that hinders clinical translation. Fluorination has proven to be a valid approach to reduce the cytotoxicity of PEI and improve the in vitro siRNA delivery potency. Hydrophobicity and lipophobicity can be controllably introduced into the side chains of PEI. However, the effect of fluorination on siRNA delivery in vivo, particularly the biodistribution of siRNA polyplex nanoparticles with fluorinated PEIs, has not been extensively explored. Here, we introduce two series of fluorinated PEIs via amidation with ethyl trifluoroacetate and perfluorobutyryl chloride. Fluorination substantially improved the performance of PEI for siRNA delivery by reducing the cytotoxicity to MDA-MB-231 cells. Importantly, fluorinated PEI enabled the major accumulation of siRNA polyplex nanoparticles in the liver while non-fluorinated PEI delivered siRNA nanoparticles mainly to the lungs after intravenous administration to mice. It is envisioned that fluorination may be an important general strategy for lowering toxicity of cationic polymers, and that the fluorination-induced alteration of biodistribution may be applicable for improved delivery to different organs. Graphical abstract.
Collapse
|
26
|
Yu X, Liu S, Cheng Q, Wei T, Lee S, Zhang D, Siegwart DJ. Lipid-Modified Aminoglycosides for mRNA Delivery to the Liver. Adv Healthc Mater 2020; 9:e1901487. [PMID: 32108440 PMCID: PMC8152636 DOI: 10.1002/adhm.201901487] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/02/2020] [Indexed: 01/10/2023]
Abstract
Cationic lipid nanoparticles (LNPs) are widely used as carriers for delivery of nucleic acids. Most synthetic routes toward cationic lipids have derived from simple amine cores. Greater chemical diversity can be obtained through starting with natural products containing basic nitrogen atoms, which offers routes to more complex molecules. Natural building blocks are not extensively explored, such as aminoglycosides, which are both structurally and functionally interesting for developing new carriers for nucleic acid delivery. Herein, cationic lipid-modified aminoglycosides (CLAs) are explored as a family of vehicles for messenger RNA (mRNA) delivery. CLAs are synthesized from natural existing aminoglycosides coupling with alkyl epoxides and acrylates. The top hit (GT-EP10) is able to deliver Luc mRNA to C57BL/6 mice at a dose of 0.05 mg kg-1 to achieve a 107 average luminescence intensity in the liver. The Lox-Stop-Lox tdTomato mouse model is used to further demonstrate that this efficient mRNA delivery system can be potentially used for gene editing. Successful delivery of human erythropoietin mRNA shows that CLA-based LNPs have promising opportunities for delivery of therapeutic nucleic acids in the future.
Collapse
Affiliation(s)
- Xueliang Yu
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shuai Liu
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qiang Cheng
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tuo Wei
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sang Lee
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Di Zhang
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Siegwart
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
27
|
Wong AH, Tran T. CD151 in Respiratory Diseases. Front Cell Dev Biol 2020; 8:64. [PMID: 32117989 PMCID: PMC7020194 DOI: 10.3389/fcell.2020.00064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/22/2020] [Indexed: 12/25/2022] Open
Abstract
The tetraspanin, Cluster of Differentiation 151 (CD151), is ubiquitously expressed in adult tissue, especially in the lungs where it has been implicated in lung cancer, asthma, influenza, and idiopathic pulmonary fibrosis (IPF). CD151 interacts with laminin-binding integrins and growth factor receptors, and is reported in cancer-promoting processes such as tumor initiation, metastasis, and angiogenesis. In asthma, CD151 was shown to promote airways hyperresponsiveness through calcium signaling whereas in influenza, CD151 was shown to be a novel host factor for nuclear viral export signaling. Furthermore, CD151 was shown to be associated with increased disease severity and poorer survival outcome in asthma and lung cancer, respectively. In this review, we provide an update on the current understanding of CD151 with regards to its contribution to lung pathophysiology. We also summarize factors that have been shown to regulate CD151 expression and identify key areas that need to be taken into consideration for its utility as a screening or prognostic tool in disease management and/or as a therapeutic target for the treatment of lung diseases.
Collapse
Affiliation(s)
- Amanda H Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
28
|
Guimaraes PPG, Zhang R, Spektor R, Tan M, Chung A, Billingsley MM, El-Mayta R, Riley RS, Wang L, Wilson JM, Mitchell MJ. Ionizable lipid nanoparticles encapsulating barcoded mRNA for accelerated in vivo delivery screening. J Control Release 2019; 316:404-417. [PMID: 31678653 PMCID: PMC7032071 DOI: 10.1016/j.jconrel.2019.10.028] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
Abstract
Messenger RNA (mRNA) has recently emerged as a promising class of nucleic acid therapy, with the potential to induce protein production to treat and prevent a range of diseases. However, the widespread use of mRNA as a therapeutic requires safe and effective in vivo delivery technologies. Libraries of ionizable lipid nanoparticles (LNPs) have been designed to encapsulate mRNA, prevent its degradation, and mediate intracellular delivery. However, these LNPs are typically characterized and screened in an in vitro setting, which may not fully replicate the biological barriers that they encounter in vivo. Here, we designed and evaluated a library of engineered LNPs containing barcoded mRNA (b-mRNA) to accelerate the screening of mRNA delivery platforms in vivo. These b-mRNA are similar in structure and function to regular mRNA, and contain barcodes that enable their delivery to be quantified via deep sequencing. Using a mini-library of b-mRNA LNPs formulated via microfluidic mixing, we show that these different formulations can be pooled together, administered intravenously into mice as a single pool, and their delivery to multiple organs (liver, spleen, brain, lung, heart, kidney, pancreas, and muscle) can be quantified simultaneously using deep sequencing. In the context of liver and spleen delivery, LNPs that exhibited high b-mRNA delivery also yielded high luciferase expression, indicating that this platform can identify lead LNP candidates as well as optimal formulation parameters for in vivo mRNA delivery. Interestingly, LNPs with identical formulation parameters that encapsulated different types of nucleic acid barcodes (b-mRNA versus a DNA barcode) altered in vivo delivery, suggesting that the structure of the barcoded nucleic acid affects LNP in vivo delivery. This platform, which enables direct barcoding and subsequent quantification of a functional mRNA, can accelerate the in vivo screening and design of LNPs for mRNA therapeutic applications such as CRISPR-Cas9 gene editing, mRNA vaccination, and other mRNA-based regenerative medicine and protein replacement therapies.
Collapse
Affiliation(s)
- Pedro P G Guimaraes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States; Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rui Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Roman Spektor
- Graduate Field of Genetics, Genomics and Development, Cornell University, Ithaca, NY, United States
| | - Mingchee Tan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Amanda Chung
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Rachel S Riley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Lili Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
29
|
Ulkoski D, Bak A, Wilson JT, Krishnamurthy VR. Recent advances in polymeric materials for the delivery of RNA therapeutics. Expert Opin Drug Deliv 2019; 16:1149-1167. [PMID: 31498013 DOI: 10.1080/17425247.2019.1663822] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The delivery of nucleic acid therapeutics through non-viral carriers face multiple biological barriers that reduce their therapeutic efficiency. Despite great progress, there remains a significant technological gap that continues to limit clinical translation of these nanocarriers. A number of polymeric materials are being exploited to efficiently deliver nucleic acids and achieve therapeutic effects. Areas covered: We discuss the recent advances in the polymeric materials for the delivery of nucleic acid therapeutics. We examine the use of common polymer architectures and highlight the challenges that exist for their development from bench side to clinic. We also provide an overview of the most notable improvements made to circumvent such challenges, including structural modification and stimuli-responsive approaches, for safe and effective nucleic acid delivery. Expert opinion: It has become apparent that a universal carrier that follows 'one-size' fits all model cannot be expected for delivery of all nucleic acid therapeutics. Carriers need to be designed to exhibit sensitivity and specificity toward individual targets diseases/indications, and relevant subcellular compartments, each of which possess their own unique challenges. The ability to devise synthetic methods that control the molecular architecture enables the future development that allow for the construction of 'intelligent' designs.
Collapse
Affiliation(s)
- David Ulkoski
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Boston , USA
| | - Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Gothenburg , Sweden
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University , Nashville , TN , USA
| | | |
Collapse
|
30
|
Lee AY, Cho MH, Kim S. Recent advances in aerosol gene delivery systems using non-viral vectors for lung cancer therapy. Expert Opin Drug Deliv 2019; 16:757-772. [PMID: 31282221 DOI: 10.1080/17425247.2019.1641083] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Lung cancer commonly occurs at a high incidence worldwide. Application of aerosol gene delivery systems using various kinds of vectors can improve the patient's quality of life by prolonging the survival rate. AREAS COVERED This review provides a recent update on aerosol gene delivery strategies using various kinds of vectors and gene-modification technologies. Peptide-mediated gene therapy achieves specific targeting of cells and highly improves efficacy. Promoter-operating expression and the CRISPR/Cas9 system are novel gene therapy strategies for effective lung cancer treatment. Furthermore, hybrid systems with a combination of vectors or drugs have been recently applied as new trends in gene therapy. EXPERT OPINION Although aerosol gene delivery has many advantages, physiological barriers in the lungs pose formidable challenges. Targeted gene delivery and gene-editing technology are promising strategies for lung cancer therapy. These strategies may allow the development of safety and high efficiency for clinical application. Recently, hybrid gene therapy combining novel and specific vectors has been developed as an advanced strategy. Although gene therapy for lung cancer is being actively researched, aerosol gene therapy strategies are currently lacking, and further studies on aerosol gene therapy are needed to treat lung cancer.
Collapse
Affiliation(s)
- Ah Young Lee
- a Center for Molecular Recognition Research, Materials and Life Science Research Division , Korea Institute of Science and Technology (KIST) , Seoul , Korea
| | - Myung-Haing Cho
- b Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine , Seoul National University , Seoul , Republic of Korea
| | - Sanghwa Kim
- c Cancer Biology Laboratory , Institut Pasteur Korea , Seongnam-si , Korea
| |
Collapse
|
31
|
Liang C, Chang J, Jiang Y, Liu J, Mao L, Wang M. Selective RNA interference and gene silencing using reactive oxygen species-responsive lipid nanoparticles. Chem Commun (Camb) 2019; 55:8170-8173. [PMID: 31241120 DOI: 10.1039/c9cc04517a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lipid-complexed small interfering RNA (siRNA) nanoparticles are promising gene regulation materials with excellent genetic, but little cellular, selectivity. Herein, we report a chemical strategy to enhance the gene silencing selectivity of these nanoparticles against cancer cells through the covalent integration of a reactive oxygen species (ROS)-degradable thioketal into the lipid nanoparticles. These lipid nanoparticles can efficiently deliver siRNA into cells, and selectively silence cancer cell gene expression in response to the high levels of intracellular ROS in cancer cells.
Collapse
Affiliation(s)
- Chunjing Liang
- Beijing National Laboratory for Molecular Science, CAS Research/Education Center for Excellence in Molecule Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Chang
- Beijing National Laboratory for Molecular Science, CAS Research/Education Center for Excellence in Molecule Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Jiang
- College of Chemistry, Beijing Normal University, Beijing 100875, China.
| | - Ji Liu
- Beijing National Laboratory for Molecular Science, CAS Research/Education Center for Excellence in Molecule Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lanqun Mao
- Beijing National Laboratory for Molecular Science, CAS Research/Education Center for Excellence in Molecule Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Wang
- Beijing National Laboratory for Molecular Science, CAS Research/Education Center for Excellence in Molecule Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
32
|
Formulation of RNA interference-based drugs for pulmonary delivery: challenges and opportunities. Ther Deliv 2019; 9:731-749. [PMID: 30277138 DOI: 10.4155/tde-2018-0029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
With recent advances in the field of RNAi-based therapeutics, it is possible to make any target gene 'druggable', at least in principle. The present review focuses on aspects critical for pulmonary delivery of formulations of nucleic acid-based drugs. The first part introduces the therapeutic potential of RNAi-based drugs for the treatment of lung diseases. Subsequently, we discuss opportunities for formulation-enabled pulmonary delivery of RNAi drugs in light of key physicochemical properties and physiological barriers. In the following section, an overview is included of methodologies for imparting inhalable characteristics to nucleic acid formulations. Finally, we review one of the bottlenecks in the early preclinical testing of inhalable nucleic acid-based formulations, in other words, devices suitable for pulmonary administration of powder-based formulations in rodents.
Collapse
|
33
|
Okuda T, Toyoda Y, Murakami T, Okamoto H. Biodistribution/biostability assessment of siRNA after intravenous and intratracheal administration to mice, based on comprehensive analysis of in vivo/ex vivo/polyacrylamide gel electrophoresis fluorescence imaging. Int J Pharm 2019; 565:294-305. [PMID: 31078647 DOI: 10.1016/j.ijpharm.2019.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/24/2019] [Accepted: 05/06/2019] [Indexed: 12/30/2022]
Abstract
We performed in vivo/ex vivo/polyacrylamide gel electrophoresis (PAGE) fluorescence imaging of near-infrared fluorescence (NIRF)-labeled siRNA (Cy5.5-siGL3) in mice to investigate the validity of each fluorescence imaging result as the biodistribution/biostability assessment of siRNA. Statistically significant correlations could be obtained between the in vivo and ex vivo fluorescence intensities of Cy5.5 in the relevant regions/tissues, except the lung region/tissue after intravenous administration. On PAGE fluorescence images with the naked formulation, there was no band corresponding to intact Cy5.5-siGL3 from all the tissues evaluated after intravenous administration, indicating that the fluorescence detected by in vivo and ex vivo fluorescence imaging was derived from degraded Cy5.5-siGL3 or free Cy5.5 cleaved from Cy5.5-siGL3. However, the band was detected from the lungs after intratracheal administration of the naked formulation, confirming higher stability of siRNA on the respiratory epithelium than in the blood. Regarding the polyethyleneimine formulation, the band was detected from all the tissues evaluated after intravenous administration and from the lungs after intratracheal administration, verifying the enhanced stability of siRNA in the body. These results clearly indicated the necessity of comprehensive analysis from in vivo/ex vivo/PAGE fluorescence imaging to precisely assess the distribution and stability of NIRF-labeled oligonucleotides including siRNA in the body.
Collapse
Affiliation(s)
- Tomoyuki Okuda
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan.
| | - Yoko Toyoda
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Takashi Murakami
- Faculty of Medicine, Saitama Medical University, 38 Moroyama, Iruma-gun, Saitama 350-0495, Japan
| | - Hirokazu Okamoto
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| |
Collapse
|
34
|
Majumder J, Taratula O, Minko T. Nanocarrier-based systems for targeted and site specific therapeutic delivery. Adv Drug Deliv Rev 2019; 144:57-77. [PMID: 31400350 PMCID: PMC6748653 DOI: 10.1016/j.addr.2019.07.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/02/2019] [Accepted: 07/05/2019] [Indexed: 01/04/2023]
Abstract
Systemic drug delivery methods such as oral or parenteral administration of free drugs possess relatively low treatment efficiency and marked adverse side effects. The use of nanoparticles for drug delivery in most cases substantially enhances drug efficacy, improves pharmacokinetics and drug release and limits their side effects. However, further enhancement in drug efficacy and significant limitation of adverse side effects can be achieved by specific targeting of nanocarrier-based delivery systems especially in combination with local administration. The present review describes major advantages and limitations of organic and inorganic nanocarriers or living cell-based drug and nucleic acid delivery systems. Among these, different nanoparticles, supramolecular gels, therapeutic cells as living drug carriers etc. have emerged as a new frontier in modern medicine.
Collapse
Affiliation(s)
- Joydeb Majumder
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; Environmental and Occupational Health Science Institute, Piscataway, NJ 08854, USA.
| |
Collapse
|
35
|
Nanotechnology in the diagnosis and treatment of lung cancer. Pharmacol Ther 2019; 198:189-205. [PMID: 30796927 DOI: 10.1016/j.pharmthera.2019.02.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 02/11/2019] [Indexed: 02/07/2023]
Abstract
Lung cancer is an umbrella term for a subset of heterogeneous diseases that are collectively responsible for the most cancer-related deaths worldwide. Despite the tremendous progress made in understanding lung tumour biology, advances in early diagnosis, multimodal therapy and deciphering molecular mechanisms of drug resistance, overall curative outcomes remain low, especially in metastatic disease. Nanotechnology, in particular nanoparticles (NPs), continue to progressively impact the way by which tumours are diagnosed and treated. The unique physicochemical properties of materials at the nanoscale grant access to a diverse molecular toolkit that can be manipulated for use in respiratory oncology. This realisation has resulted in several clinically approved NP formulations and many more in clinical trials. However, NPs are not a panacea and have yet to be utilised to maximal effect in lung cancer, and medicine in a wider context. This review serves to: describe the complexity of lung cancer, the current diagnostic and therapeutic environment, and highlight the recent advancements of nanotechnology based approaches in diagnosis and treatment of respiratory malignancies. Finally, a brief outlook on the future directions of nanomedicine is provided; presently the full potential of the field is yet to be realised. By gleaning lessons and integrating advancements from neighbouring disciplines, nanomedicine can be elevated to a position where the current barriers that stymie full clinical impact are lifted.
Collapse
|
36
|
Cheng Q, Wei T, Jia Y, Farbiak L, Zhou K, Zhang S, Wei Y, Zhu H, Siegwart DJ. Dendrimer-Based Lipid Nanoparticles Deliver Therapeutic FAH mRNA to Normalize Liver Function and Extend Survival in a Mouse Model of Hepatorenal Tyrosinemia Type I. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1805308. [PMID: 30368954 DOI: 10.1002/adma.201805308] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/14/2018] [Indexed: 06/08/2023]
Abstract
mRNA-mediated protein replacement represents a promising concept for the treatment of liver disorders. Children born with fumarylacetoacetate hydrolase (FAH) mutations suffer from Hepatorenal Tyrosinemia Type 1 (HT-1) resulting in renal dysfunction, liver failure, neurological impairments, and cancer. Protein replacement therapy using FAH mRNA offers tremendous potential to cure HT-1, but is currently hindered by the development of effective mRNA carriers that can function in diseased livers. Structure-guided, rational optimization of 5A2-SC8 mRNA-loaded dendrimer lipid nanoparticles (mDLNPs) increases delivery potency of FAH mRNA, resulting in functional FAH protein and sustained normalization of body weight and liver function in FAH-/- knockout mice. Optimization using luciferase mRNA produces DLNP carriers that are efficacious at mRNA doses as low as 0.05 mg kg-1 in vivo. mDLNPs transfect > 44% of all hepatocytes in the liver, yield high FAH protein levels (0.5 mg kg-1 mRNA), and are well tolerated in a knockout mouse model with compromised liver function. Genetically engineered FAH-/- mice treated with FAH mRNA mDLNPs have statistically equivalent levels of TBIL, ALT, and AST compared to wild type C57BL/6 mice and maintain normal weight throughout the month-long course of treatment. This study provides a framework for the rational optimization of LNPs to improve delivery of mRNA broadly and introduces a specific and viable DLNP carrier with translational potential to treat genetic diseases of the liver.
Collapse
Affiliation(s)
- Qiang Cheng
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tuo Wei
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yuemeng Jia
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lukas Farbiak
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kejin Zhou
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shuyuan Zhang
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yonglong Wei
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
37
|
Alhajj N, Chee CF, Wong TW, Rahman NA, Abu Kasim NH, Colombo P. Lung cancer: active therapeutic targeting and inhalational nanoproduct design. Expert Opin Drug Deliv 2018; 15:1223-1247. [DOI: 10.1080/17425247.2018.1547280] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Nasser Alhajj
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Chin Fei Chee
- Nanotechnology & Catalysis Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Noorsaadah Abd Rahman
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- Wellness Research Cluster, Institute of Research Management & Monitoring, University of Malaya, Kuala Lumpur, Malaysia
| | - Paolo Colombo
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| |
Collapse
|
38
|
Polyester-based nanoparticles for nucleic acid delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:983-994. [DOI: 10.1016/j.msec.2018.07.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
|
39
|
Magalhães M, Alvarez-Lorenzo C, Concheiro A, Figueiras A, Santos AC, Veiga F. RNAi-based therapeutics for lung cancer: biomarkers, microRNAs, and nanocarriers. Expert Opin Drug Deliv 2018; 15:965-982. [PMID: 30232915 DOI: 10.1080/17425247.2018.1517744] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Despite the current advances in the discovery of the lung cancer biomarkers and, consequently, in the diagnosis, this pathology continues to be the primary cause of cancer-related death worldwide. In most cases, the illness is diagnosed in an advanced stage, which limits the current treatment options available and reduces the survival rate. Therefore, RNAi-based therapy arises as a promising option to treat lung cancer. AREAS COVERED This review provides an overview on the exploitation of lung cancer biology to develop RNAi-based therapeutics to be applied in the treatment of lung cancer. Furthermore, the review analyzes the main nanocarriers designed to deliver RNAi molecules and induce antitumoral effects in lung cancer, and provides updated information about current RNAi-based therapeutics for lung cancer in clinical trials. EXPERT OPINION RNAi-based therapy uses nanocarriers to perform a targeted and efficient delivery of therapeutic genes into lung cancer cells, by taking advantage of the known biomarkers in lung cancer. These therapeutic genes are key regulatory molecules of crucial cellular pathways involved in cell proliferation, migration, and apoptosis. Thereby, the characteristics and functionalization of the nanocarrier and the knowledge of lung cancer biology have direct influence in improving the therapeutic effect of this therapy.
Collapse
Affiliation(s)
- Mariana Magalhães
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal
| | - Carmen Alvarez-Lorenzo
- c Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+D Pharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS) , Universidade de Santiago de Compostela , Santiago de Compostela , Spain
| | - Angel Concheiro
- c Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+D Pharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS) , Universidade de Santiago de Compostela , Santiago de Compostela , Spain
| | - Ana Figueiras
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal
| | - Ana Cláudia Santos
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal
| | - Francisco Veiga
- a Department of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal.,b REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy , University of Coimbra , Coimbra , Portugal
| |
Collapse
|
40
|
|
41
|
Liu S, Jia H, Yang J, Pan J, Liang H, Zeng L, Zhou H, Chen J, Guo T. Zinc Coordinated Cationic Polymers Break Up the Paradox between Low Molecular Weight and High Transfection Efficacy. Biomacromolecules 2018; 19:4270-4276. [DOI: 10.1021/acs.biomac.8b01140] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Shuai Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Huiting Jia
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Jixiang Yang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Jianping Pan
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Huiyun Liang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Liheng Zeng
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Hao Zhou
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Jiatong Chen
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Tianying Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
42
|
Liu S, Yang J, Jia H, Zhou H, Chen J, Guo T. Virus Spike and Membrane-Lytic Mimicking Nanoparticles for High Cell Binding and Superior Endosomal Escape. ACS APPLIED MATERIALS & INTERFACES 2018; 10:23630-23637. [PMID: 29931973 DOI: 10.1021/acsami.8b06934] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Virus-inspired mimics for gene therapy have attracted increasing attention because viral vectors show robust efficacy owing to the highly infectious nature and efficient endosomal escape. Nonetheless, until now, synthetic materials have failed to achieve high "infectivity," and especially, the mimicking of virus spikes for "infection" is underappreciated. Herein, a virus spike mimic by a zinc (Zn) coordinative ligand that shows high affinity toward phosphate-rich cell membranes is reported. Surprisingly, this ligand also demonstrates superior functionality of destabilizing endosomes. Therefore, the Zn coordination is more likely to imitate the virus nature with high cell binding and endosomal membrane disruption. Following this, the Zn coordinative ligand is functionalized on a bioreducible cross-linked peptide with alkylation that imitates the viral lipoprotein shell. The ultimate virus-mimicking nanoparticle closely imitates the structures and functions of viruses, leading to robust transfection efficiency both in vitro and in vivo. More importantly, apart from targeting ligand- and cell-penetrating peptide, the metal coordinative ligand may provide another option to functionalize diverse biomaterials for enhanced efficacy, demonstrating its broad referential significance to pursue nonviral vectors with high performance.
Collapse
|
43
|
Liu S, Jia H, Yang J, Pan J, Liang H, Zeng L, Zhou H, Chen J, Guo T. Zinc Coordination Substitute Amine: A Noncationic Platform for Efficient and Safe Gene Delivery. ACS Macro Lett 2018; 7:868-874. [PMID: 35650761 DOI: 10.1021/acsmacrolett.8b00374] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Amines have been extensively involved in vector design thus far, however, their clinical translation has been impeded by several obstacles: cytotoxicity, polyplex serum instability and low efficacy in vivo. In pursuit of functional groups to substitute amines in vector design to address these disadvantages is of great significance. Herein, we report well-tailored noncationic copolymers that contain hydrophilic, hydrophobic, and zinc coordinative moieties through reversible addition-fragmentation chain transfer (RAFT) polymerization for efficient and safe gene delivery. These polymers are capable of condensing DNA, enabling the formation of uncharged polyplexes. Especially, the zinc coordinative ligand can simultaneously benefit strong DNA binding, robust cellular uptake, efficacious endosomal destabilization, low cytotoxicity, and avoidance of serum protein adsorption. The coordinative module holds great promise to substitute amines and inspires the development of next-generation gene vectors. More importantly, the coordinative copolymers illuminate the possibility and potential of noncationic gene delivery systems for clinical applications.
Collapse
Affiliation(s)
- Shuai Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Huiting Jia
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Jixiang Yang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Jianping Pan
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Huiyun Liang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Liheng Zeng
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Hao Zhou
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Jiatong Chen
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Tianying Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
44
|
Far-red light-mediated programmable anti-cancer gene delivery in cooperation with photodynamic therapy. Biomaterials 2018; 171:72-82. [DOI: 10.1016/j.biomaterials.2018.04.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/05/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022]
|
45
|
Abstract
Polymeric nanoparticles have tremendous potential to improve the efficacy of therapeutic cancer treatments by facilitating targeted delivery to a desired site. The physical and chemical properties of polymers can be tuned to accomplish delivery across the multiple biological barriers required to reach diverse subsets of cells. The use of biodegradable polymers as nanocarriers is especially attractive, as these materials can be designed to break down in physiological conditions and engineered to exhibit triggered functionality when at a particular location or activated by an external source. We present how biodegradable polymers can be engineered as drug delivery systems to target the tumor microenvironment in multiple ways. These nanomedicines can target cancer cells directly, the blood vessels that supply the nutrients and oxygen that support tumor growth, and immune cells to promote anticancer immunotherapy.
Collapse
Affiliation(s)
- Johan Karlsson
- Department of Biomedical Engineering, Translational Tissue Engineering Center, and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA;
- Department of Chemistry, Ångström Laboratory, Uppsala University, Uppsala SE-75121, Sweden
| | - Hannah J Vaughan
- Department of Biomedical Engineering, Translational Tissue Engineering Center, and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA;
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center, and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA;
- Departments of Materials Science and Engineering, Chemical and Biomolecular Engineering, Neurosurgery, Oncology, and Ophthalmology and the Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| |
Collapse
|
46
|
Li J, Liang H, Liu J, Wang Z. Poly (amidoamine) (PAMAM) dendrimer mediated delivery of drug and pDNA/siRNA for cancer therapy. Int J Pharm 2018; 546:215-225. [PMID: 29787895 DOI: 10.1016/j.ijpharm.2018.05.045] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022]
Abstract
Poly (amidoamine) (PAMAM) dendrimers are well-defined, highly branched macromolecules with numerous active amine groups on the surface. Because of their unique properties, PAMAM dendrimers have steadily grown in popularity in drug delivery, gene therapy, medical imaging and diagnostic application. This review focuses on the recent developments on the application in PAMAM dendrimers as effective carriers for drug and gene (pDNA, siRNA) delivery in cancer therapy, including: a) PAMAM for anticancer drug delivery; b) PAMAM and gene therapy; c) PAMAM used in overcoming tumor multidrug resistance; d) PAMAM used for hybrid nanoparticles; and e) PAMAM linked or loaded in other nanoparticles.
Collapse
Affiliation(s)
- Jun Li
- School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| | - Huamin Liang
- Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230088, Anhui, China
| | - Jing Liu
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing 100084, China
| | - Ziyuan Wang
- School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| |
Collapse
|
47
|
Xiong H, Zhou K, Yan Y, Miller JB, Siegwart DJ. Tumor-Activated Water-Soluble Photosensitizers for Near-Infrared Photodynamic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:16335-16343. [PMID: 29697248 DOI: 10.1021/acsami.8b04710] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Current photosensitizers (PSs) for photodynamic therapy (PDT) are limited by their low water solubility and tendency to aggregate, low near-infrared (NIR) absorption, and low cancer selectivity. Here, we designed iodinated, water-soluble NIR boron dipyrromethene-based PSs to achieve image-guided and efficient PDT against cancer in vivo that is enhanced by leveraging tumor-specific pH-responsive activation. PEG2k5c-I and PEG2k5c-OMe-I localized to tumors and were activated by acidic pH in the tumor microenvironment to produce 1O2 and fluorescence for efficient PDT and effective cancer detection after intravenous administration. Upon NIR irradiation, these PSs exhibited strong NIR absorption at 660 and 690 nm, stable NIR emission at 692 and 742 nm, and high 1O2 quantum yields of 0.78 and 0.72 in acidic pH. PEG2k5c-I and PEG2k5c-OMe-I killed cancer cells upon irradiation of NIR light and were nontoxic without irradiation. Light-activated PDT treatment of breast cancer tumors in mice resulted in suppression of tumor growth, DNA damage, and necrosis selectively in tumors. This work thus introduces a versatile method to directly synthesize modular pH-responsive water-soluble PSs and provides a versatile strategy for activatable PDT against cancer.
Collapse
Affiliation(s)
- Hu Xiong
- Department of Biochemistry, Simmons Comprehensive Cancer Center , The University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Kejin Zhou
- Department of Biochemistry, Simmons Comprehensive Cancer Center , The University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Yunfeng Yan
- Department of Biochemistry, Simmons Comprehensive Cancer Center , The University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Jason B Miller
- Department of Biochemistry, Simmons Comprehensive Cancer Center , The University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Daniel J Siegwart
- Department of Biochemistry, Simmons Comprehensive Cancer Center , The University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| |
Collapse
|
48
|
Guan X, Chang Y, Sun J, Song J, Xie Y. Engineered Hsp Protein Nanocages for siRNA Delivery. Macromol Biosci 2018; 18:e1800013. [DOI: 10.1002/mabi.201800013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/24/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Xingang Guan
- Life Science Research Center; Beihua University; Jilin 132013 P. R. China
| | - Yu Chang
- Life Science Research Center; Beihua University; Jilin 132013 P. R. China
- College of Medicine; Beihua University; Jilin 132013 P. R. China
| | - Jinghui Sun
- College of Medicine; Beihua University; Jilin 132013 P. R. China
| | - Jianxi Song
- College of Medicine; Beihua University; Jilin 132013 P. R. China
| | - Yu Xie
- Life Science Research Center; Beihua University; Jilin 132013 P. R. China
| |
Collapse
|
49
|
Development of spray-freeze-dried siRNA/PEI powder for inhalation with high aerosol performance and strong pulmonary gene silencing activity. J Control Release 2018; 279:99-113. [PMID: 29627404 DOI: 10.1016/j.jconrel.2018.04.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/21/2018] [Accepted: 04/03/2018] [Indexed: 11/20/2022]
Abstract
In the present study, a novel dry small interfering RNA (siRNA) powder for inhalation, containing polyethyleneimine (PEI) as a delivery vector, was produced by spray freeze drying (SFD). The powder had spherical and highly porous structure of approximately 10 μm in diameter with high aerosol performance for emission and lung delivery. The reconstituted siRNA/PEI complex after dissolution of the powder had almost the same physicochemical properties and in vitro gene silencing activity as the original one constituted in the sample solution before SFD, showing that the integrity of the siRNA was maintained. In in vivo studies of intratracheal administration into lung metastasis mice and healthy mice, powder with a low dose of 3 μg siRNA exhibited strong and specific gene silencing activity against tumors metastasized to the lungs, whereas it caused no significant histological changes, lactate dehydrogenase leakage, or inflammatory cytokine induction in the lungs. These results strongly indicated that inhalable dry siRNA/PEI powders can provide effective pulmonary gene silencing without severe lung injury and that SFD can be applied to the production of such powders.
Collapse
|
50
|
Miller JB, Kos P, Tieu V, Zhou K, Siegwart DJ. Development of Cationic Quaternary Ammonium Sulfonamide Amino Lipids for Nucleic Acid Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:2302-2311. [PMID: 29286232 DOI: 10.1021/acsami.7b15982] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Lipid nanoparticles (LNPs) currently comprise the most effective carrier class for the delivery of small RNAs. Among lipid carriers, charge-unbalanced lipids are relatively unexplored synthetically. Herein, we developed and evaluated a novel collection of compounds for small interfering RNA (siRNA) delivery, termed cationic quaternary ammonium sulfonamide amino lipids (CSALs). The formulated CSAL LNPs containing cholesterol, 1,2-distearoyl-sn-glycero-3-phosphocholine, and lipid poly(ethylene glycol) exhibited biophysical property trends directly related to the CSAL chemical structure. Lead CSAL LNPs were identified using an siRNA delivery screen. Further chemical synthesis using a rational structure-guided design showed that the head group structure could alter the pKa and other physical properties that modulated delivery efficacy. Shorter-chained dimethylamino head groups, acetate side chains, and higher tail carbon numbers were favorable for delivery. This led to a further study of A3-OAc-C2Me LNPs, which enabled in vivo delivery to normal mouse lungs and subcutaneous and orthotopic lung tumors. Incorporation of CSALs into liver-targeting formulations shifted the in vivo delivery of these carriers to the lungs. This study highlights the importance of the cationic lipid structure in LNPs and provides further design guidelines for nucleic acid carriers.
Collapse
Affiliation(s)
- Jason B Miller
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Petra Kos
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Victor Tieu
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Kejin Zhou
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| |
Collapse
|