1
|
Luo X, Weidinger E, Burghardt T, Höhn M, Wagner E. CRISPR/Cas9 Ribonucleoprotein Delivery Enhanced by Lipo-Xenopeptide Carriers and Homology-Directed Repair Modulators: Insights from Reporter Cell Lines. Int J Mol Sci 2025; 26:4361. [PMID: 40362595 PMCID: PMC12073011 DOI: 10.3390/ijms26094361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
CRISPR-Cas9 genome editing is a versatile platform for studying and treating various diseases. Homology-directed repair (HDR) with DNA donor templates serves as the primary pathway for gene correction in therapeutic applications, but its efficiency remains a significant challenge. This study investigates strategies to enhance gene correction efficiency using a T-shaped lipo-xenopeptide (XP)-based Cas9 RNP/ssDNA delivery system combined with various HDR enhancers. Nu7441, a known DNA-PKcs inhibitor, was found to be most effective in enhancing HDR-mediated gene correction. An over 10-fold increase in HDR efficiency was achieved by Nu7441 in HeLa-eGFPd2 cells, with a peak HDR efficiency of 53% at a 5 nM RNP concentration and up to 61% efficiency confirmed by Sanger sequencing. Surprisingly, the total gene editing efficiency including non-homologous end joining (NHEJ) was also improved. For example, Nu7441 boosted exon skipping via NHEJ-mediated splice site destruction by 30-fold in a DMD reporter cell model. Nu7441 modulated the cell cycle by reducing cells in the G1 phase and extending the S and G2/M phases without compromising cellular uptake or endosomal escape. The enhancement in genome editing by Nu7441 was widely applicable across several cell lines, several Cas9 RNP/ssDNA carriers (LAF-XPs), and also Cas9 mRNA/sgRNA/ssDNA polyplexes. These findings highlight a novel and counterintuitive role for Nu7441 as an enhancer of both HDR and total gene editing efficiency, presenting a promising strategy for Cas9 RNP-based gene therapy.
Collapse
Affiliation(s)
- Xianjin Luo
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (X.L.); (E.W.); (T.B.); (M.H.)
| | - Eric Weidinger
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (X.L.); (E.W.); (T.B.); (M.H.)
| | - Tobias Burghardt
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (X.L.); (E.W.); (T.B.); (M.H.)
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (X.L.); (E.W.); (T.B.); (M.H.)
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (X.L.); (E.W.); (T.B.); (M.H.)
- Center for Nanoscience (CeNS), LMU Munich, 80799 Munich, Germany
- CNATM—Cluster for Nucleic Acid Therapeutics Munich, 81377 Munich, Germany
| |
Collapse
|
2
|
Korleski J, Sall S, Luly KM, Johnson MK, Johnson AL, Khela H, Lal B, Taylor TC, Ashby JM, Alonso H, Li A, Zhou W, Smith-Connor K, Hughes R, Tzeng SY, Laterra J, Green JJ, Lopez-Bertoni H. Multipronged SMAD pathway targeting by lipophilic poly(β-amino ester) miR-590-3p nanomiRs inhibits mesenchymal glioblastoma growth and prolongs survival. Signal Transduct Target Ther 2025; 10:145. [PMID: 40301302 PMCID: PMC12041600 DOI: 10.1038/s41392-025-02223-w] [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: 09/17/2024] [Revised: 02/25/2025] [Accepted: 03/27/2025] [Indexed: 05/01/2025] Open
Abstract
ASBSTRACT Despite aggressive therapy, glioblastoma (GBM) recurs in almost all patients and treatment options are very limited. Despite our growing understanding of how cellular transitions associate with relapse in GBM, critical gaps remain in our ability to block these molecular changes and treat recurrent disease. In this study we combine computational biology, forward-thinking understanding of miRNA biology and cutting-edge nucleic acid delivery vehicles to advance targeted therapeutics for GBM. Computational analysis of RNA sequencing from clinical GBM specimens identified TGFβ type II receptor (TGFBR2) as a key player in the mesenchymal transition associated with worse outcome in GBM. Mechanistically, we show that elevated levels of TGFBR2 is conducive to reduced temozolomide (TMZ) sensitivity. This effect is, at least partially, induced by stem-cell driving events coordinated by the reprogramming transcription factors Oct4 and Sox2 that lead to open chromatin states. We show that blocking TGFBR2 via molecular and pharmacological approaches decreases stem cell capacity and sensitivity of clinical recurrent GBM (rGBM) isolates to TMZ in vitro. Network analysis uncovered miR-590-3p as a tumor suppressor that simultaneously inhibits multiple oncogenic nodes downstream of TGFBR2. We also developed novel biodegradable lipophilic poly(β-amino ester) nanoparticles (LiPBAEs) for in vivo microRNA (miRNAs) delivery. Following direct intra-tumoral infusion, these nanomiRs efficiently distribute through the tumors. Importantly, miR-590-3p nanomiRs inhibited the growth and extended survival of mice bearing orthotopic human rGBM xenografts, with an apparent 30% cure rate. These results show that miRNA-based targeted therapeutics provide new opportunities to treat rGBM and bypass the resistance to standard of care therapy.
Collapse
Affiliation(s)
- Jack Korleski
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota, USA
| | - Sophie Sall
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
| | - Kathryn M Luly
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Maya K Johnson
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Biology, Johns Hopkins University, Baltimore, USA
| | - Amanda L Johnson
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Harmon Khela
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, USA
| | - Bachchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - T C Taylor
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, USA
| | - Jean Micheal Ashby
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Hector Alonso
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
| | - Alice Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
| | - Weiqiang Zhou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | | | - Russell Hughes
- Single Cell & Transcriptomics Core at the Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jordan J Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Departments of Materials Science & Engineering and Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, USA.
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, USA.
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, USA.
| | - Hernando Lopez-Bertoni
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, USA.
| |
Collapse
|
3
|
Liu C, Tian X, Wang Z, Mak JCW, Mao S, Liu TM, Zheng Y. Hydrogen-induced disruption of the airway mucus barrier enhances nebulized RNA delivery to reverse pulmonary fibrosis. SCIENCE ADVANCES 2025; 11:eadt2752. [PMID: 40238879 PMCID: PMC12002117 DOI: 10.1126/sciadv.adt2752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Nebulized RNA therapies are well suited for treating respiratory diseases, in particular pulmonary fibrosis (PF); however, effective delivery remains challenging. In this study, we present a highly efficient aerosol inhalation system that enables high levels of in vivo transfection efficiency in lung macrophages, yielding durable responses against PF. First, we established a nose-only aerosol inhalation device integrated with a hydrogen supplement system. This setup enables the precise administration of lipid nanoparticles (LNPs) at a controlled low dose, while simultaneously delivering the optimal concentration of therapeutic hydrogen gas. We further developed a hybrid lipid NP (HNP) by hybridizing a pH-dependent charge-inverting lipid film with apoptotic T cell membranes to enhance endosomal escape and trigger macrophage production of hepatocyte growth factor for lung repair. We demonstrated that the hydrogen flow-induced shear stresses disrupt the NP-mucus interaction, enhancing the deposition of aerosolized HNPs/TGFβ1 siRNA within fibrotic lung lesions, effectively blocking fibrogenic signaling pathways and offering a clinically viable strategy for combating PF.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xidong Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Judith Choi Wo Mak
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences & Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau 999078, China
| |
Collapse
|
4
|
Shao S, Chen X, Chen Y, Wang M, Huang J, Wang J, Yu X, Li X, Zhang Y, Ping Y, Hu Q, Liang T. STING-activating pyroptotic nanoparticles for in situ pancreatic cancer vaccination and immunotherapy. Sci Bull (Beijing) 2025:S2095-9273(25)00364-0. [PMID: 40253299 DOI: 10.1016/j.scib.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/17/2025] [Accepted: 03/25/2025] [Indexed: 04/21/2025]
Affiliation(s)
- Shiyi Shao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China
| | - Xiaohong Chen
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China
| | - Yuxuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310030, China
| | - Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China
| | - Junmin Huang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China
| | - Jianwei Wang
- Department of Chemistry, Zhejiang University, Hangzhou 310030, China
| | - Xiazhen Yu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China
| | - Xinya Li
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China
| | - Yao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310030, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310030, China
| | - Qida Hu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China.
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou 310003, China; MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Innovation Center for the Study of Pancreatic Diseases, Hangzhou 310003, China; Zhejiang University Cancer Center, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
5
|
Klipp A, Greitens C, Scherer D, Elsener A, Leroux J, Burger M. Modular Calcium-Responsive and CD9-Targeted Phospholipase System Enhancing Endosomal Escape for DNA Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410815. [PMID: 39998318 PMCID: PMC12005733 DOI: 10.1002/advs.202410815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/12/2025] [Indexed: 02/26/2025]
Abstract
Gene delivery systems must overcome multiple barriers, with endosomal escape representing a prominent obstacle. We have previously shown that a bacterial phospholipase C (PLC) enabled endosomal escape of a non-viral protein-based DNA delivery system termed TFAMoplex. Building upon this, this work introduces a calcium-responsive system designed to enhance endosomal escape through non-covalent capturing of PLC to the TFAMoplex followed by its release within endosomes and nanobody-mediated targeting to the endosomal membrane. This approach leads to improved TFAMoplexes enabling transfection of HeLa cells in full serum with a half maximal effective concentration (EC50) of less than 200 ng DNA per mL serum, using only 5 nM PLC. Particularly, the modular capture, release and targeting system could potentially be adapted to other delivery agents previously constrained by poor endosomal escape. These findings present a promising strategy to achieve efficient endosomal escape, offering prospects for improved delivery of macromolecules, in particular nucleic acids.
Collapse
Affiliation(s)
- Alexander Klipp
- Department of Chemistry and Applied BiosciencesETH ZürichVladimir‐Prelog‐Weg 3Zürich8093Switzerland
| | - Christina Greitens
- Department of Chemistry and Applied BiosciencesETH ZürichVladimir‐Prelog‐Weg 3Zürich8093Switzerland
| | - David Scherer
- Department of Chemistry and Applied BiosciencesETH ZürichVladimir‐Prelog‐Weg 3Zürich8093Switzerland
| | - Alexander Elsener
- Department of Chemistry and Applied BiosciencesETH ZürichVladimir‐Prelog‐Weg 3Zürich8093Switzerland
| | - Jean‐Christophe Leroux
- Department of Chemistry and Applied BiosciencesETH ZürichVladimir‐Prelog‐Weg 3Zürich8093Switzerland
| | - Michael Burger
- Department of Chemistry and Applied BiosciencesETH ZürichVladimir‐Prelog‐Weg 3Zürich8093Switzerland
| |
Collapse
|
6
|
He X, Wang R, Cao Y, Ding Y, Chang Y, Dong H, Xie R, Zhong G, Yang H, Li J. Lung-Specific mRNA Delivery by Ionizable Lipids with Defined Structure-Function Relationship and Unique Protein Corona Feature. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416525. [PMID: 39965058 PMCID: PMC11984862 DOI: 10.1002/advs.202416525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/31/2025] [Indexed: 02/20/2025]
Abstract
Targeted delivery of mRNA with lipid nanoparticles (LNPs) holds great potential for treating pulmonary diseases. However, the lack of rational design principles for efficient lung-homing lipids hinders the prevalence of mRNA therapeutics in this organ. Herein, the combinatorial screening with structure-function analysis is applied to rationalize the design strategy for nonpermanently charged lung-targeted ionizable lipids. It is discovered that lipids carrying N-methyl and secondary amine groups in the heads, and three tails originated from epoxyalkanes, exhibiting superior pulmonary selectivity and efficiency. Representative ionizable lipids with systematically variation in chemical structures are selected to study the well-known but still puzzling "protein corona" adsorbed on the surface of LNPs. In addition to the commonly used corona-biomarker vitronectin, other arginine-glycine-aspartic acid (RGD)-rich proteins usually involved in collagen-containing extracellular matrix, such as fibrinogen and fibronectin have also been identified to have a strong correlation with lung tropism. This work provides insight into the rational design of lung-targeting ionizable lipids and reveals a previously unreported potential function of RGD-rich proteins in the protein corona of lung-homing LNPs.
Collapse
Affiliation(s)
- Xiaoyan He
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
| | - Runyuan Wang
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
| | - Yan Cao
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
| | - Yan Ding
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
| | - Yan Chang
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
| | - Haoru Dong
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghai200040China
| | - Rong Xie
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghai200040China
| | - Guisheng Zhong
- iHuman InstituteShanghaiTech UniversityShanghai201210China
| | - Huiying Yang
- Department of PharmacyHuashan HospitalFudan UniversityShanghai200040China
| | - Jianfeng Li
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
| |
Collapse
|
7
|
Gilbert BR, Miglani C, Karmakar A, Pal M, Chandran VC, Gupta S, Pal A, Ganguli M. A combination of systemic mannitol and mannitol modified polyester nanoparticles for caveolae-mediated gene delivery to the brain. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102480. [PMID: 40104113 PMCID: PMC11919422 DOI: 10.1016/j.omtn.2025.102480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/04/2025] [Indexed: 03/20/2025]
Abstract
Overcoming the blood-brain barrier (BBB) remains a significant challenge for nucleic acid delivery to the brain. We have explored a combination of mannitol-modified poly (β-amino ester) (PBAE) nanoparticles and systemic mannitol injection for crossing the BBB. We incorporated mannitol in the PBAE polymer for caveolae targeting and selected monomers that may help avoid delivery to the liver. We also induced caveolae at the BBB through systemic mannitol injection in order to create an opportunity for the caveolae-targeting nanoparticles (M30 D90) containing plasmid DNA to cross the BBB. When a clinically relevant dose was administered intravenously in this caveolae induction model, M30 D90 demonstrated significant transgene expression of a reporter plasmid in the brain, with selective uptake by neuronal cells and minimal liver accumulation. We demonstrate that caveolae modulation using systemic mannitol administration and caveolae targeting using designed nanoparticles are necessary for efficient delivery to the brain. This delivery platform offers a simple, scalable, and controlled delivery solution and holds promise for treating brain diseases with functional targets.
Collapse
Affiliation(s)
- Betsy Reshma Gilbert
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chirag Miglani
- Chemical Biology Unit, Institute of Nanoscience and Technology, Sector 81, Mohali, Punjab 140306, India
| | | | - Muneesh Pal
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vysakh C Chandran
- Chemical Biology Unit, Institute of Nanoscience and Technology, Sector 81, Mohali, Punjab 140306, India
| | - Sarika Gupta
- National Institute of Immunology, New Delhi 110067, India
| | - Asish Pal
- Chemical Biology Unit, Institute of Nanoscience and Technology, Sector 81, Mohali, Punjab 140306, India
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
8
|
Fatima M, An T, Hong KJ. Revolutionizing mRNA Vaccines Through Innovative Formulation and Delivery Strategies. Biomolecules 2025; 15:359. [PMID: 40149895 PMCID: PMC11940278 DOI: 10.3390/biom15030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Modernization of existing methods for the delivery of mRNA is vital in advanced therapeutics. Traditionally, mRNA has faced obstacles of poor stability due to enzymatic degradation. This work examines cutting-edge formulation and emerging techniques for safer delivery of mRNA vaccines. Inspired by the success of lipid nanoparticles (LNP) in delivering mRNA vaccines for COVID-19, a variety of other formulations have been developed to deliver mRNA vaccines for diverse infections. The meritorious features of nanoparticle-based mRNA delivery strategies, including LNP, polymeric, dendrimers, polysaccharide-based, peptide-derived, carbon and metal-based, DNA nanostructures, hybrid, and extracellular vesicles, have been examined. The impact of these delivery platforms on mRNA vaccine delivery efficacy, protection from enzymatic degradation, cellular uptake, controlled release, and immunogenicity has been discussed in detail. Even with significant developments, there are certain limitations to overcome, including toxicity concerns, limited information about immune pathways, the need to maintain a cold chain, and the necessity of optimizing administration methods. Continuous innovation is essential for improving delivery systems for mRNA vaccines. Future research directions have been proposed to address the existing challenges in mRNA delivery and to expand their potential prophylactic and therapeutic application.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Timothy An
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
9
|
Weiss S, Decker S, Kugler C, Gómez LB, Fasching H, Benisch D, Alioglu F, Ferencz L, Birkfeld T, Ilievski F, Baumann V, Duran A, Dusinovic E, Follrich N, Milenkovic S, Mihalicokova D, Paunov D, Singeorzan K, Zehetmayer N, Zivanonvic D, Lächelt U, Boersma A, Rülicke T, Sami H, Ogris M. Accelerated Endosomal Escape of Splice-Switching Oligonucleotides Enables Efficient Hepatic Splice Correction. ACS APPLIED MATERIALS & INTERFACES 2025; 17:9000-9018. [PMID: 39873730 PMCID: PMC11826512 DOI: 10.1021/acsami.4c19340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/01/2025] [Accepted: 01/14/2025] [Indexed: 01/30/2025]
Abstract
Splice-switching oligonucleotides (SSOs) can restore protein functionality in pathologies and are promising tools for manipulating the RNA-splicing machinery. Delivery vectors can considerably improve SSO functionality in vivo and allow dose reduction, thereby addressing the challenges of RNA-targeted therapeutics. Here, we report a biocompatible SSO nanocarrier, based on redox-responsive disulfide cross-linked low-molecular-weight linear polyethylenimine (cLPEI), for overcoming multiple biological barriers from subcellular compartments to en-route serum stability and finally in vivo delivery challenges. Intracellularly responsive cross-links of cLPEI significantly accelerated the endosomal escape and offered efficient SSO release to the cell's nucleus, thereby leading to high splice correction in vitro. In vivo performance of cLPEI-SSOs was investigated in a novel transgenic mouse model for splice correction, spatiotemporal tracking of SSO delivery in wild-type mice, and biodistribution in a colorectal cancer peritoneal metastasis model. A single intravenous application of 5 mg kg-1 cLPEI-SSOs induced splice correction in liver, lung, kidney, and bladder, giving functional protein, which was validated by RT-PCR. Near-infrared (NIR) fluorescence imaging and X-ray computed tomography revealed improved organ retention and reduced renal excretion of SSOs. NIR microscopy demonstrated the accumulation of SSOs in angiogenic tumors within the pancreas. Successful nuclear delivery of SSOs was observed in the hepatocytes. Thus, cLPEI nanocarriers resulted in highly efficient splice correction in vivo, highlighting the critical role of the enhanced SSO bioavailability.
Collapse
Affiliation(s)
- Silvia Weiss
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Simon Decker
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Christoph Kugler
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Laura Bocanegra Gómez
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Helene Fasching
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Denise Benisch
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Fatih Alioglu
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Levente Ferencz
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Theresa Birkfeld
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Filip Ilievski
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Volker Baumann
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Alina Duran
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Enes Dusinovic
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Nadine Follrich
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Sandra Milenkovic
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Dajana Mihalicokova
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Daniel Paunov
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Karla Singeorzan
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Nikolaus Zehetmayer
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Dejan Zivanonvic
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Ulrich Lächelt
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Auke Boersma
- Institute
of In-Vivo and In-Vitro Models, Biomodels Austria, Department of Biomedical
Sciences, University of Veterinary Medicine
Vienna, Veterinärplatz
1, A-1210 Vienna, Austria
| | - Thomas Rülicke
- Department
of Biomedical Sciences and Pathobiology, University of Veterinary Medicine Vienna and Ludwig Boltzmann Institute
for Hematology and Oncology, Veterinärplatz 1, A-1210 Vienna, Austria
| | - Haider Sami
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Manfred Ogris
- Faculty
of Life Sciences, Department of Pharmaceutical Sciences, Laboratory
of Macromolecular Cancer Therapeutics (MMCT), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| |
Collapse
|
10
|
Bader J, Rüedi P, Mantella V, Geisshüsler S, Brigger F, Qureshi BM, Ortega Arroyo J, Montanari E, Leroux J. Loading of Extracellular Vesicles with Nucleic Acids via Hybridization with Non-Lamellar Liquid Crystalline Lipid Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404860. [PMID: 39741121 PMCID: PMC11848734 DOI: 10.1002/advs.202404860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 11/01/2024] [Indexed: 01/02/2025]
Abstract
The translation of cell-derived extracellular vesicles (EVs) into biogenic gene delivery systems is limited by relatively inefficient loading strategies. In this work, the loading of various nucleic acids into small EVs via their spontaneous hybridization with preloaded non-lamellar liquid crystalline lipid nanoparticles (LCNPs), forming hybrid EVs (HEVs) is described. It is demonstrated that LCNPs undergo pH-dependent structural transitions from inverse hexagonal (HII) phases at pH 5 to more disordered non-lamellar phases, possibly inverse micellar (L2) or sponge (L3) phases, at pH 7.4, which are particularly suitable for inducing a controlled hybridization process with EVs. State-of-the-art single-particle analysis techniques reveal that LCNPs interact with various EV subpopulations at physiological conditions and that ≈40% of HEVs are loaded with the genetic cargo. Importantly, this study demonstrates that EV membrane proteins remain accessible on HEV surfaces, with their intrinsic enzymatic activity unaffected after the hybridization process. Finally, HEVs show in vitro improved transfection efficiencies compared to unhybridized LCNPs. In summary, this versatile platform holds potential for loading various nucleic acid molecules into native EVs and may help developing EV-based therapeutics.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Pascal Rüedi
- Nanophotonic Systems LaboratoryDepartment of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
| | - Valeria Mantella
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Silvana Geisshüsler
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Bilal Muhammad Qureshi
- Scientific Center for Optical and Electron Microscopy (ScopeM)ETH ZurichZurich8093Switzerland
| | - Jaime Ortega Arroyo
- Nanophotonic Systems LaboratoryDepartment of Mechanical and Process EngineeringETH ZurichZurich8092Switzerland
| | - Elita Montanari
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Jean‐Christophe Leroux
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| |
Collapse
|
11
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
12
|
Luo X, Germer J, Burghardt T, Grau M, Lin Y, Höhn M, Lächelt U, Wagner E. Dual pH-responsive CRISPR/Cas9 ribonucleoprotein xenopeptide complexes for genome editing. Eur J Pharm Sci 2025; 205:106983. [PMID: 39647515 DOI: 10.1016/j.ejps.2024.106983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR associated (Cas) protein has been proved as a powerful tool for the treatment of genetic diseases. The Cas9 protein, when combined with single-guide RNA (sgRNA), forms a Cas9/sgRNA ribonucleoprotein (RNP) capable of targeting and editing the genome. However, the limited availability of effective carriers has restricted the broader application of CRISPR/Cas9 RNP. In this study, we evaluated dual pH-responsive amphiphilic xenopeptides (XPs) for delivering CRISPR/Cas9 RNP. These artificial lipo-XPs contain apolar cationizable lipoamino fatty acid (LAF) and polar cationizable oligoaminoethylene acid units such as succinoyl-tetraethylenepentamine (Stp) in various ratios and U-shaped topologies. The carriers were screened for functional Cas9/sgRNA RNP delivery in four different reporter cell lines, including a Duchenne muscular dystrophy (DMD) exon skipping reporter cell model. Significantly enhanced cellular uptake into HeLa cells, effective endosomal disruption in HeLa gal8-mRuby3 cells, and potent genome editing by several Cas9/sgRNA RNP complexes was observed in four different cell lines in the 5 nM sgRNA range. Comparing Cas9/sgRNA RNP complexes with Cas9 mRNA/sgRNA polyplexes in the DMD reporter cell model demonstrated similar splice site editing and high exon skipping of the two different molecular Cas9 modalities. Based on these studies, analogues of two potent U1 LAF2-Stp and LAF4-Stp2 structures were deployed, tuning the amphiphilicity of the polar Stp group by replacement with the six oligoamino acids dmGtp, chGtp, dGtp, Htp, Stt, or GEIPA. The most potent LAF2-Stp analogues (containing dGtp, chGtp or GEIPA) demonstrated further enhanced gene editing efficiency with EC50 values of 1 nM in the DMD exon skipping reporter cell line. Notably, the EC50 of LAF2-dGtp reached 0.51 nM even upon serum incubation. Another carrier (LAF4-GEIPA2) complexing Cas9/sgRNA RNP and donor DNA, facilitated up to 43 % of homology-directed repair (HDR) in HeLa eGFPd2 cells visualized by the switch from green fluorescent protein (eGFP) to blue fluorescent protein (BFP). This study presents a delivery system tunable for Cas9 RNP complexes or Cas9 RNP/donor DNA polyplexes, offering an effective and easily applicable strategy for gene editing.
Collapse
Affiliation(s)
- Xianjin Luo
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Janin Germer
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Tobias Burghardt
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Melina Grau
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Yi Lin
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ulrich Lächelt
- Center for Nanoscience (CeNS), LMU Munich, 80799 Munich, Germany; Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; Center for Nanoscience (CeNS), LMU Munich, 80799 Munich, Germany; CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany.
| |
Collapse
|
13
|
Austria E, Bilek M, Varamini P, Akhavan B. Breaking biological barriers: Engineering polymeric nanoparticles for cancer therapy. NANO TODAY 2025; 60:102552. [DOI: 10.1016/j.nantod.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Mancino C, Franke M, Greco A, Sontam T, Mcculloch P, Corbo C, Taraballi F. RNA therapies for musculoskeletal conditions. J Control Release 2025; 377:756-766. [PMID: 39617171 DOI: 10.1016/j.jconrel.2024.11.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024]
Abstract
Musculoskeletal conditions impact 1.71 billion individuals, posing significant challenges due to their complexity, varying clinical courses, and unclear molecular mechanisms. Conventional spectrum treatments often prove inadequate, underscoring the importance of targeted therapies. Recently, RNA-based technologies have emerged as a groundbreaking approach in therapeutics, showing applications in joint related ailments. This perspective aims to examine endeavors exploring the use of RNA-based treatments in both experimental and clinical contexts for addressing joint issues like osteoarthritis, rheumatoid arthritis, and cartilage injuries. The cited studies demonstrate how mRNA can stimulate the production of proteins that aid in controlling inflammation, fostering tissue regeneration and repairing cartilage damage. In summary, this perspective offers an overview of the progress made in mRNA-based technologies for treating related conditions by highlighting favorable findings from preclinical research and encouraging results from clinical trials. With advancements in the field, mRNA therapeutics have the potential to revolutionize treatment approaches for musculoskeletal disorders, bringing renewed hope to the future of musculoskeletal conditions.
Collapse
Affiliation(s)
- Chiara Mancino
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Madeline Franke
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
| | - Antonietta Greco
- School of Medicine and Surgery, Nanomedicine Center Nanomib, University of Milano-Bicocca, Via R. Follereau 3, 20854 Vedano al Lambro, MB, Italy
| | - Tarun Sontam
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
| | - Patrick Mcculloch
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Claudia Corbo
- School of Medicine and Surgery, Nanomedicine Center Nanomib, University of Milano-Bicocca, Via R. Follereau 3, 20854 Vedano al Lambro, MB, Italy; IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20161 Milan, Italy.
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA; Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
15
|
Yu M, Lin L, Zhou D, Liu S. Interaction design in mRNA delivery systems. J Control Release 2025; 377:413-426. [PMID: 39580076 DOI: 10.1016/j.jconrel.2024.11.038] [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: 10/11/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Following the coronavirus disease 2019 (COVID-19) pandemic, mRNA technology has made significant breakthroughs, emerging as a potential universal platform for combating various diseases. To address the challenges associated with mRNA delivery, such as instability and limited delivery efficacy, continuous advancements in genetic engineering and nanotechnology have led to the exploration and refinement of various mRNA structural modifications and delivery platforms. These achievements have significantly broadened the clinical applications of mRNA therapies. Despite the progress, the understanding of the interactions in mRNA delivery systems remains limited. These interactions are complex and multi-dimensional, occurring between mRNA and vehicles as well as delivery materials and helper ingredients. Resultantly, stability of the mRNA delivery systems and their delivery efficiency can be both significantly affected. This review outlines the current state of mRNA delivery strategies and summarizes the interactions in mRNA delivery systems. The interactions include the electrostatic interactions, hydrophobic interactions, hydrogen bonding, π-π stacking, coordination interactions, and so on. This interaction understanding provides guideline for future design of next-generation mRNA delivery systems, thereby offering new perspectives and strategies for developing diverse mRNA therapeutics.
Collapse
Affiliation(s)
- Mengyao Yu
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Lixin Lin
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Shuai Liu
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
16
|
Wang H, Cheng Y. Polymers for mRNA Delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70002. [PMID: 39763235 DOI: 10.1002/wnan.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/22/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
mRNA delivery has emerged as a transformative approach in biotechnology and medicine, offering a versatile platform for the development of novel therapeutics. Unlike traditional small molecule drugs or protein-based biologics, mRNA therapeutics have the unique ability to direct cells to generate therapeutic proteins, allowing for precise modulation of biological processes. The delivery of mRNA into target cells is a critical step in realizing the therapeutic potential of this technology. In this review, our focus is on the latest advancements in designing functional polymers to achieve efficient mRNA delivery. Biodegradable polymers and low molecular weight polymers in addressing the balance in mRNA binding and release are summarized. Benefiting from the excellent performance of lipid nanoparticles in mRNA delivery, polymer/lipid hybrid nanostructures are also included. Finally, the challenges and future prospects in the development of polymer-based mRNA delivery systems are discussed.
Collapse
Affiliation(s)
- Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
17
|
Lim Y, Campochiaro PA, Green JJ. Suprachoroidal Delivery of Viral and Nonviral Vectors for Treatment of Retinal and Choroidal Vascular Diseases. Am J Ophthalmol 2024:S0002-9394(24)00571-3. [PMID: 39716546 DOI: 10.1016/j.ajo.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024]
Abstract
PURPOSE Current treatments for retinal and choroidal neovascular diseases suffer from insufficient durability, including anti-vascular endothelial growth factor-A agents. It is, therefore, of interest to explore alternative methods that could allow for robust improvement in visual acuity with fewer injections required. DESIGN Literature review. RESULTS Among various preclinical and clinical studies in the literature, a promising approach is the use of suprachoroidal injection with viral and nonviral gene delivery vectors. Compared with other ocular injection methods, suprachoroidal injection has demonstrated wide biodistribution of injected agents and safety as an outpatient procedure. In terms of viral vectors, suprachoroidal injection of an adeno-associated virus 8 vector expressing an anti-vascular endothelial growth factor-A antibody fragment has shown an excellent safety profile and evidence of biological activity. In terms of nonviral vectors, lipid nanoparticles and polymeric nanoparticles both demonstrate strong promise for ocular gene therapy in large animal models. In particular, biodegradable poly(β-amino ester) nanoparticles show excellent biodistribution, safety, and efficacy for gene therapy via the suprachoroidal route. Nonviral nanoparticle approaches can have notable advantages over viral vectors in terms of carrying capacity, redosability, and manufacturing costs. An advantage of gene therapy is that once a delivery vector has been optimized, genetic cargos can be readily tailored without changing the safety, efficacy, and pharmacokinetic properties of the delivery vector. CONCLUSIONS This review highlights recent progress that has been made and compares viral and nonviral suprachoroidal gene delivery for the treatment of retinal and choroidal vascular diseases. Suprachoroidal gene therapy is an emerging biotechnology that holds substantial potential to make a translational impact in treating these diseases.
Collapse
Affiliation(s)
- Yeongseo Lim
- From the Department of Biomedical Engineering (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center (Y.L., J.J.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Johns Hopkins Translational ImmunoEngineering Center (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter A Campochiaro
- Department of Ophthalmology (P.A.C., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Department of Neuroscience (P.A.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Jordan J Green
- From the Department of Biomedical Engineering (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center (Y.L., J.J.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Johns Hopkins Translational ImmunoEngineering Center (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Department of Ophthalmology (P.A.C., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Departments of Chemical & Biomolecular Engineering and Materials Science & Engineering (J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Departments of Neurosurgery and Oncology (J.J.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Institute for Nanobiotechnology, Johns Hopkins University (J.J.G.), Baltimore, Maryland, USA..
| |
Collapse
|
18
|
Li Q, Byun J, Kim D, Wu Y, Lee J, Oh YK. Cell membrane-coated mRNA nanoparticles for enhanced delivery to dendritic cells and immunotherapy. Asian J Pharm Sci 2024; 19:100968. [PMID: 39640052 PMCID: PMC11617980 DOI: 10.1016/j.ajps.2024.100968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/30/2024] [Accepted: 07/24/2024] [Indexed: 12/07/2024] Open
Abstract
Cationic polymers such as polyethylenimine have been considered promising carriers for mRNA vaccines. However, their application is hindered by their inherent toxicity and a lack of targeted delivery capability. These issues need to be addressed to develop effective cancer vaccines. In this study, we investigated whether dendritic cell membrane-coated polyethylenimine/mRNA nanoparticles (DPN) could effectively deliver mRNA to dendritic cells and induce immune responses. For comparison, we employed red blood cell membrane-coated polyethylenimine/mRNA (RPN) and plain polyethylenimine/mRNA polyplex (PN). The dendritic cell membrane coating altered the zeta potential values and surface protein patterns of PN. DPN demonstrated significantly higher uptake in dendritic cells compared to PN and RPN, and it also showed greater mRNA expression within these cells. DPN, carrying mRNA encoding luciferase, enhanced green fluorescent protein, or ovalbumin (OVA), exhibited higher protein expression in dendritic cells than the other groups. Additionally, DPN exhibited favorable mRNA escape from lysosomes post-internalization into dendritic cells. In mice, subcutaneous administration of DPN containing ovalbumin mRNA (DPNOVA) elicited higher titers of anti-OVA IgG antibodies and a greater population of OVA-specific CD8+ T cells than the other groups. In a B16F10-OVA tumor model, DPNOVA treatment resulted in the lowest tumor growth among the treated groups. Moreover, the population of OVA-specific CD8+ T cells was the highest in the DPNOVA-treated group. While we demonstrated DPN's feasibility as an mRNA delivery system in a tumor model, the potential of DPN can be broadly extended for immunotherapeutic treatments of various diseases through mRNA delivery to antigen-presenting cells.
Collapse
Affiliation(s)
| | | | - Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
19
|
Bernstein ZJ, Gierke TR, Dammen-Brower K, Tzeng SY, Zhu S, Chen SS, Wilson DS, Green JJ, Yarema KJ, Spangler JB. Production of site-specific antibody conjugates using metabolic glycoengineering and novel Fc glycovariants. J Biol Chem 2024; 300:108005. [PMID: 39551135 PMCID: PMC11697773 DOI: 10.1016/j.jbc.2024.108005] [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: 07/25/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Molecular conjugation to antibodies has emerged as a growing strategy to combine the mechanistic activities of the attached molecule with the specificity of antibodies. A variety of technologies have been applied for molecular conjugation; however, these approaches face several limitations, including disruption of antibody structure, destabilization of the antibody, and/or heterogeneous conjugation patterns. Collectively, these challenges lead to reduced yield, purity, and function of conjugated antibodies. While glycoengineering strategies have largely been applied to study protein glycosylation and manipulate cellular metabolism, these approaches also harbor great potential to enhance the production and performance of protein therapeutics. Here, we devise a novel glycoengineering workflow for the development of site-specific antibody conjugates. This approach combines metabolic glycoengineering using azido-sugar analogs with newly installed N-linked glycosylation sites in the antibody constant domain to achieve specific conjugation to the antibody via the introduced N-glycans. Our technique allows facile and efficient manufacturing of well-defined antibody conjugates without the need for complex or destructive chemistries. Moreover, the introduction of conjugation sites in the antibody fragment crystallizable (Fc) domain renders this approach widely applicable and target agnostic. Our platform can accommodate up to three conjugation sites in tandem, and the extent of conjugation can be tuned through the use of different sugar analogs or production in different cell lines. We demonstrated that our platform is compatible with various use-cases, including fluorescent labeling, antibody-drug conjugation, and targeted gene delivery. Overall, this study introduces a versatile and effective yet strikingly simple approach to producing antibody conjugates for research, industrial, and medical applications.
Collapse
Affiliation(s)
- Zachary J Bernstein
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Taylor R Gierke
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kris Dammen-Brower
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Stanley Zhu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sabrina S Chen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - D Scott Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin J Yarema
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Molecular Microbiology & Immunology, Johns Hopkins University School of Public Health, Baltimore, Maryland, USA.
| |
Collapse
|
20
|
Rocher EE, Luly KM, Tzeng SY, Sunshine JC, Green JJ. Efficient Polymeric Nanoparticle Gene Delivery Enabled Via Tri- and Tetrafunctional Branching. Biomacromolecules 2024; 25:7260-7273. [PMID: 39466232 DOI: 10.1021/acs.biomac.4c00954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Poly(β-amino ester) (PBAE) nanoparticles (NPs) show great promise for nonviral gene delivery. Recent studies suggest branched PBAEs (BPBAEs) offer advantages over linear counterparts, but the effect of polymer structure has not been well investigated across many chemical constituents. Here, a library of BPBAEs was synthesized with tri- and tetrafunctional branching. These polymers self-assemble with DNA to form highly cationic, monodisperse NPs with notably small size (∼50 nm). Optimal transfection occurred with polymer structures that featured moderate PBAE branching, enabling complete DNA encapsulation, rapid NP uptake, and robust expression at low DNA doses and polymer amounts. Optimized NPs enabled efficient DNA delivery to diverse cell types in vitro while maintaining high cellular viability, demonstrating significant improvements over a well-performing linear PBAE counterpart. BPBAEs also facilitated efficient mRNA and siRNA delivery, highlighting the versatility of these structures and demonstrating the broad utility of BPBAE NPs as vectors for nucleic acid delivery.
Collapse
|
21
|
Pant A, Jain A, Chen Y, Patel K, Saleh L, Tzeng S, Nitta RT, Zhao L, Wu CYJ, Bederson M, Wang WL, Bergsneider BHL, Choi J, Medikonda R, Verma R, Cho KB, Kim LH, Kim JE, Yazigi E, Lee SY, Rajendran S, Rajappa P, Mackall CL, Li G, Tyler B, Brem H, Pardoll DM, Lim M, Jackson CM. The CCR6-CCL20 Axis Promotes Regulatory T-cell Glycolysis and Immunosuppression in Tumors. Cancer Immunol Res 2024; 12:1542-1558. [PMID: 39133127 DOI: 10.1158/2326-6066.cir-24-0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/20/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Regulatory T cells (Treg) are important players in the tumor microenvironment. However, the mechanisms behind their immunosuppressive effects are poorly understood. We found that CCR6-CCL20 activity in tumor-infiltrating Tregs is associated with greater glycolytic activity and ablation of Ccr6 reduced glycolysis and lactic acid production while increasing compensatory glutamine metabolism. Immunosuppressive activity toward CD8+ T cells was abrogated in Ccr6-/- Tregs due to reduction in activation-induced glycolysis. Furthermore, Ccr6-/- mice exhibited improved survival across multiple tumor models compared to wild-type mice and Treg and CD8+ T-cell depletion abrogated the improvement. In addition, Ccr6 ablation further promoted the efficacy of anti-PD-1 therapy in a preclinical glioma model. Follow-up knockdown of Ccl20 with siRNA also demonstrated improvement in antitumor efficacy. Our results unveil CCR6 as a marker and regulator of Treg-induced immunosuppression and identify approaches to target the metabolic determinants of Treg immunosuppressive activity.
Collapse
Affiliation(s)
- Ayush Pant
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aanchal Jain
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yiyun Chen
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, California
| | - Kisha Patel
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laura Saleh
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stephany Tzeng
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Ryan T Nitta
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Liang Zhao
- Department of Oncology and Medicine, Bloomberg-Kimmel Institute for Immunotherapy, the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Maria Bederson
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - William Lee Wang
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, California
| | | | - John Choi
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Ravi Medikonda
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Rohit Verma
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Kwang Bog Cho
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Lily H Kim
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Jennifer E Kim
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eli Yazigi
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Si Yeon Lee
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Sakthi Rajendran
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Prajwal Rajappa
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Crystal L Mackall
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, California
| | - Gordon Li
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Betty Tyler
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henry Brem
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Drew M Pardoll
- Department of Oncology and Medicine, Bloomberg-Kimmel Institute for Immunotherapy, the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Christopher M Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
22
|
Al‐Kharboosh R, Bechtle A, Tzeng SY, Zheng J, Mondal SK, Wilson DR, Perez‐Vega C, Green JJ, Quiñones‐Hinojosa A. Therapeutic potential and impact of nanoengineered patient-derived mesenchymal stem cells in a murine resection and recurrence model of human glioblastoma. Bioeng Transl Med 2024; 9:e10675. [PMID: 39545093 PMCID: PMC11558202 DOI: 10.1002/btm2.10675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 11/17/2024] Open
Abstract
Confounding results of engineered mesenchymal stem cells (MSCs) used as cellular vehicles has plagued technologies whereby success or failure of novel approaches may be dismissed or inaccurately ascribed solely to the biotechnology platform rather than suitability of the human donor. Polymeric materials were screened for non-viral engineering of MSCs from multiple human donors to deliver bone morphogenic protein-4 (BMP4), a protein previously investigated in clinical trials for glioblastoma (GBM) to combat a subpopulation of highly invasive and tumorigenic clones. A "smart technology" that target the migratory and stem-like nature of GBM will require: (1) a cellular vehicle (MSC) which can scavenge and target residual cells left behind after surgical debulking and deliver; (2) anti-glioma cargo (BMP4). Multiple MSC donors are safely engineered, though varied in susceptibility to accept BMP4 due to intrinsic characteristics revealed by their molecular signatures. Efficiency is compared via secretion, downstream signaling, differentiation, and anti-proliferative properties across all donors. In a clinically relevant resection and recurrence model of patient-derived human GBM, we demonstrate that nanoengineered MSCs are not "donor agnostic" and efficacy is influenced by the inherent suitability of the MSC to the cargo. Therefore, donor profiles hold greater influence in determining downstream outcomes than the technical capabilities of the engineering technology.
Collapse
Affiliation(s)
- Rawan Al‐Kharboosh
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
- Department of NeuroscienceMayo Clinic Graduate SchoolJacksonvilleFloridaUSA
- AtPoint tx Co.WashingtonDistrict of ColumbiaUSA
| | - Alex Bechtle
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
| | - Stephany Y. Tzeng
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jiaying Zheng
- Department of NeuroscienceMayo Clinic Graduate SchoolJacksonvilleFloridaUSA
| | | | - David R. Wilson
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Jordan J. Green
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Departments of Neurosurgery, Oncology, Ophthalmology, Materials Science & Engineering, and Chemical & Biomolecular EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | |
Collapse
|
23
|
Oude Egberink R, van Schie DM, Joosten B, de Muynck LTA, Jacobs W, van Oostrum J, Brock R. Unraveling mRNA delivery bottlenecks of ineffective delivery vectors by co-transfection with effective carriers. Eur J Pharm Biopharm 2024; 202:114414. [PMID: 39009193 DOI: 10.1016/j.ejpb.2024.114414] [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: 05/25/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
The messenger RNA (mRNA) SARS-CoV-2 vaccines have demonstrated the therapeutic potential of this novel drug modality. Protein expression is the consequence of a multistep delivery process that relies on proper packaging into nanoparticle carriers to protect the mRNA against degradation enabling effective cellular uptake and endosomal release, and liberating the mRNA in the cytosol. Bottlenecks along this route remain challenging to pinpoint. Although methods to assess endosomal escape of carriers have been developed, versatile strategies to identify bottlenecks along the delivery trajectory are missing. Here, it is shown that co-incubating an inefficient nanoparticle formulation with an efficient one solves this problem. Cells were co-incubated with mRNA nanoparticles formed with either the efficient cell-penetrating peptide (CPP) PepFect14 or the inefficient CPP nona-arginine (R9). Co-transfection enhanced cellular uptake and endosomal escape of R9-formulated mRNA, resulting in protein expression, demonstrating that both vectors enter cells along the same route. In addition, cells were transfected with a galectin-9-mCherry fusion protein to detect endosomal rupture. Remarkably, despite endosomal release, mRNA remained confined to punctate structures, identifying mRNA liberation as a further bottleneck. In summary, co-transfection offers a rapid means to identify bottlenecks in cytosolic mRNA delivery, supporting the rational design and optimization of intracellular mRNA delivery systems.
Collapse
Affiliation(s)
- Rik Oude Egberink
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| | - Deni M van Schie
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| | - Ben Joosten
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| | - Lisa T A de Muynck
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| | - Ward Jacobs
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| | - Jenny van Oostrum
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| | - Roland Brock
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain.
| |
Collapse
|
24
|
Hu Y, Tzeng SY, Cheng L, Lin J, Villabona-Rueda A, Yu S, Li S, Schneiderman Z, Zhu Y, Ma J, Wilson DR, Shannon SR, Warren T, Rui Y, Qiu C, Kavanagh EW, Luly KM, Zhang Y, Korinetz N, D’Alessio FR, Wang TH, Kokkoli E, Reddy SK, Luijten E, Green JJ, Mao HQ. Supramolecular assembly of polycation/mRNA nanoparticles and in vivo monocyte programming. Proc Natl Acad Sci U S A 2024; 121:e2400194121. [PMID: 39172792 PMCID: PMC11363337 DOI: 10.1073/pnas.2400194121] [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: 01/04/2024] [Accepted: 07/19/2024] [Indexed: 08/24/2024] Open
Abstract
Size-dependent phagocytosis is a well-characterized phenomenon in monocytes and macrophages. However, this size effect for preferential gene delivery to these important cell targets has not been fully exploited because commonly adopted stabilization methods for electrostatically complexed nucleic acid nanoparticles, such as PEGylation and charge repulsion, typically arrest the vehicle size below 200 nm. Here, we bridge the technical gap in scalable synthesis of larger submicron gene delivery vehicles by electrostatic self-assembly of charged nanoparticles, facilitated by a polymer structurally designed to modulate internanoparticle Coulombic and van der Waals forces. Specifically, our strategy permits controlled assembly of small poly(β-amino ester)/messenger ribonucleic acid (mRNA) nanoparticles into particles with a size that is kinetically tunable between 200 and 1,000 nm with high colloidal stability in physiological media. We found that assembled particles with an average size of 400 nm safely and most efficiently transfect monocytes following intravenous administration and mediate their differentiation into macrophages in the periphery. When a CpG adjuvant is co-loaded into the particles with an antigen mRNA, the monocytes differentiate into inflammatory dendritic cells and prime adaptive anticancer immunity in the tumor-draining lymph node. This platform technology offers a unique ligand-independent, particle-size-mediated strategy for preferential mRNA delivery and enables therapeutic paradigms via monocyte programming.
Collapse
Affiliation(s)
- Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Leonardo Cheng
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Jinghan Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Andres Villabona-Rueda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Shuai Yu
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL60208
| | - Sixuan Li
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Zachary Schneiderman
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Jingyao Ma
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - David R. Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Sydney R. Shannon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Tiarra Warren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Yuan Rui
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Chenhu Qiu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Erin W. Kavanagh
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Kathryn M. Luly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Yicheng Zhang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Nicole Korinetz
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Franco R. D’Alessio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Tza-Huei Wang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Efrosini Kokkoli
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Sashank K. Reddy
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Erik Luijten
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL60208
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL60208
- Department of Chemistry, Northwestern University, Evanston, IL60208
- Department of Physics and Astronomy, Northwestern University, Evanston, IL60208
| | - Jordan J. Green
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
25
|
Jogdeo CM, Siddhanta K, Das A, Ding L, Panja S, Kumari N, Oupický D. Beyond Lipids: Exploring Advances in Polymeric Gene Delivery in the Lipid Nanoparticles Era. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404608. [PMID: 38842816 PMCID: PMC11384239 DOI: 10.1002/adma.202404608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/23/2024] [Indexed: 06/07/2024]
Abstract
The recent success of gene therapy during the COVID-19 pandemic has underscored the importance of effective and safe delivery systems. Complementing lipid-based delivery systems, polymers present a promising alternative for gene delivery. Significant advances have been made in the recent past, with multiple clinical trials progressing beyond phase I and several companies actively working on polymeric delivery systems which provides assurance that polymeric carriers can soon achieve clinical translation. The massive advantage of structural tunability and vast chemical space of polymers is being actively leveraged to mitigate shortcomings of traditional polycationic polymers and improve the translatability of delivery systems. Tailored polymeric approaches for diverse nucleic acids and for specific subcellular targets are now being designed to improve therapeutic efficacy. This review describes the recent advances in polymer design for improved gene delivery by polyplexes and covalent polymer-nucleic acid conjugates. The review also offers a brief note on novel computational techniques for improved polymer design. The review concludes with an overview of the current state of polymeric gene therapies in the clinic as well as future directions on their translation to the clinic.
Collapse
Affiliation(s)
- Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ashish Das
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
26
|
Grau M, Wagner E. Strategies and mechanisms for endosomal escape of therapeutic nucleic acids. Curr Opin Chem Biol 2024; 81:102506. [PMID: 39096817 DOI: 10.1016/j.cbpa.2024.102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 08/05/2024]
Abstract
Despite impressive recent establishment of therapeutic nucleic acids as drugs and vaccines, their broader medical use is impaired by modest performance in intracellular delivery. Inefficient endosomal escape presents a major limitation responsible for inadequate cytosolic cargo release. Depending on the carrier, this endosomal barrier can strongly limit or even abolish nucleic acid delivery. Different recent endosomal escape strategies and their hypothesized mechanisms are reviewed.
Collapse
Affiliation(s)
- Melina Grau
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany; Center for Nanoscience (CeNS), LMU Munich, 80799 Munich, Germany.
| |
Collapse
|
27
|
Steffens RC, Thalmayr S, Weidinger E, Seidl J, Folda P, Höhn M, Wagner E. Modulating efficacy and cytotoxicity of lipoamino fatty acid nucleic acid carriers using disulfide or hydrophobic spacers. NANOSCALE 2024; 16:13988-14005. [PMID: 38984864 DOI: 10.1039/d4nr01357c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Double pH-responsive xenopeptides comprising polar ionizable succinoyl tetraethylene pentamine (Stp) motifs and lipophilic ionizable lipoamino fatty acids (LAFs) were recently found to efficiently transfect mRNA and pDNA at low doses. However, potency was often accompanied with cytotoxicity at higher doses. Insertion of bioreducible disulfide building blocks (ssbb) or non-reducible hydrophobic spacers between polar and apolar ionizable domains of LAF-Stp carriers should mitigate toxicity of xenopeptides. Carriers showed stable nucleic acid complexation and endosomal pH-dependent lytic activities, both of which were abolished after reductive cleavage of ssbb-containing carriers. For pDNA, U-shaped carriers with one Stp and two LAF units or bundle carriers with two Stps and four LAFs displayed highest potency. For mRNA, best transfection was achieved with bundle carriers with one Stp and four LAFs. Both the ssbb and hydrophobic spacer containing analogs displayed improved metabolic activity, reduced membrane damage, and improved cell growth. The ssbb carriers were most beneficial regarding living cell count and low apoptosis rates. Mechanistically, inserted spacers decelerated the transfection kinetics and altered the requirement of endosomal protonation. Overall, mRNA and pDNA carriers with improved biocompatibility have been designed, with their high potency illustrated in transfection of various cell lines including low passage number colon carcinoma cells.
Collapse
Affiliation(s)
- Ricarda C Steffens
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Sophie Thalmayr
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany
| | - Eric Weidinger
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
| | - Johanna Seidl
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany
| | - Paul Folda
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
- CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany
| |
Collapse
|
28
|
Kromer AE, Sieber-Schäfer F, Farfan Benito J, Merkel OM. Design of Experiments Grants Mechanistic Insights into the Synthesis of Spermine-Containing PBAE Copolymers. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37545-37554. [PMID: 38985802 PMCID: PMC11284743 DOI: 10.1021/acsami.4c06079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024]
Abstract
Successful therapeutic delivery of siRNA with polymeric nanoparticles seems to be a promising but not vastly understood and complicated goal to achieve. Despite years of research, no polymer-based delivery system has been approved for clinical use. Polymers, as a delivery system, exhibit considerable complexity and variability, making their consistent production a challenging endeavor. However, a better understanding of the polymerization process of polymer excipients may improve the reproducibility and material quality for more efficient use in drug products. Here, we present a combination of Design of Experiment and Python-scripted data science to establish a prediction model, from which important parameters can be extracted that influence the synthesis results of polybeta-amino esters (PBAEs), a common type of polymer used preclinically for nucleic acid delivery. We synthesized a library of 27 polymers, each one at different temperatures with different reaction times and educt ratios using an orthogonal central composite (CCO-) design. This design allowed a detailed characterization of factor importance and interactions using a very limited number of experiments. We characterized the polymers by analyzing the resulting composition by 1H-NMR and the size distribution by GPC measurements. To further understand the complex mechanism of block polymerization in a one-pot synthesis, we developed a Python script that helps us to understand possible step-growth steps. We successfully developed and validated a predictive response surface and gathered a deeper understanding of the synthesis of polyspermine-based amphiphilic PBAEs.
Collapse
Affiliation(s)
- Adrian
P. E. Kromer
- Pharmaceutical
Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich 81377, Germany
| | - Felix Sieber-Schäfer
- Pharmaceutical
Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich 81377, Germany
| | - Johan Farfan Benito
- Pharmaceutical
Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich 81377, Germany
- Université
Paris Cité, Paris 75015, France
| | - Olivia M. Merkel
- Pharmaceutical
Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich 81377, Germany
- Center
for NanoScience Munich (CeNS), Munich 81377, Germany
- Cluster
for Nucleic Acid Therapeutics Munich (CNATM), Munich 81377, Germany
| |
Collapse
|
29
|
Yong H, Lin L, Li Z, Guo R, Wang C, Liu S, Zhou D. Tailoring Highly Branched Poly(β-amino ester)s for Efficient and Organ-Selective mRNA Delivery. NANO LETTERS 2024. [PMID: 39013032 DOI: 10.1021/acs.nanolett.4c02440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Development of mRNA therapeutics necessitates targeted delivery technology, while the clinically advanced lipid nanoparticles face difficulty for extrahepatic delivery. Herein, we design highly branched poly(β-amino ester)s (HPAEs) for efficacious organ-selective mRNA delivery through tailoring their chemical compositions and topological structures. Using an "A2+B3+C2" Michael addition platform, a combinatorial library of 219 HPAEs with varied backbone structures, terminal groups, and branching degrees are synthesized. The branched topological structures of HPAEs provide enhanced serum resistance and significantly higher mRNA expression in vivo. The terminal amine structures of HPAEs determine the organ-selectivity of mRNA delivery following systemic administration: morpholine facilitates liver targeting, ethylenediamine favors spleen delivery, while methylpentane enables mRNA delivery to the liver, spleen, and lungs simultaneously. This study represents a comprehensive exploration of the structure-activity relationship governing both the efficiency and organ-selectivity of mRNA delivery by HPAEs, suggesting promising candidates for treating various organ-related diseases.
Collapse
Affiliation(s)
- Haiyang Yong
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Lixin Lin
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Zhejiang University, Hangzhou 310058, China
| | - Zhili Li
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Rui Guo
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Chenfei Wang
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, Liangzhu Laboratory, Zhejiang University, Hangzhou 310058, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
30
|
Shang L, Jiang X, Zhao X, Huang X, Wang X, Jiang X, Kong X, Yao M, Jiang S, Wong PP. Mitochondrial DNA-boosted dendritic cell-based nanovaccination triggers antitumor immunity in lung and pancreatic cancers. Cell Rep Med 2024; 5:101648. [PMID: 38986624 PMCID: PMC11293323 DOI: 10.1016/j.xcrm.2024.101648] [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: 11/23/2023] [Revised: 04/09/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
Low migratory dendritic cell (DC) levels pose a challenge in cancer immune surveillance, yet their impact on tumor immune status and immunotherapy responses remains unclear. We present clinical evidence linking reduced migratory DC levels to immune-cold tumor status, resulting in poor patient outcomes. To address this, we develop an autologous DC-based nanovaccination strategy using patient-derived organoid or cancer cell lysate-pulsed cationic nanoparticles (cNPs) to load immunogenic DC-derived microvesicles (cNPcancer cell@MVDC). This approach transforms immune-cold tumors, increases migratory DCs, activates T cells and natural killer cells, reduces tumor growth, and enhances survival in orthotopic pancreatic and lung cancer models, surpassing conventional methods. In vivo imaging reveals superior cNPcancer cell@MVDC accumulation in tumors and lymph nodes, promoting immune cell infiltration. Mechanistically, cNPs enrich mitochondrial DNA, enhancing cGAS-STING-mediated DC activation and migration. Our strategy shifts cold tumors to a hot state, enhancing antitumor immunity for potential personalized cancer treatments.
Collapse
Affiliation(s)
- Lihuan Shang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xue Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xinbao Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xi Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaojuan Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xue Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Mingkang Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Respiratory Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shanping Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Respiratory Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ping-Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
31
|
Tiwade PB, Ma Y, VanKeulen-Miller R, Fenton OS. A Lung-Expressing mRNA Delivery Platform with Tunable Activity in Hypoxic Environments. J Am Chem Soc 2024; 146:17365-17376. [PMID: 38874565 DOI: 10.1021/jacs.4c04565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Messenger RNA (mRNA) delivery platforms often facilitate protein expression in the liver following intravenous injection and have been optimized for use in normally oxygenated cells (21% O2 atmosphere). However, there is a growing need for mRNA therapy in diseases affecting non-liver organs, such as the lungs. Additionally, many diseases are characterized by hypoxia (<21% O2 atmosphere), a state of abnormally low oxygenation in cells and tissues that can reduce the efficacy of mRNA therapies by upwards of 80%. Here, we report a Tunable Lung-Expressing Nanoparticle Platform (TULEP) for mRNA delivery, whose properties can be readily tuned for optimal expression in hypoxic environments. Briefly, our study begins with the synthesis and characterization of a novel amino acrylate polymer that can be effectively complexed with mRNA payloads into TULEPs. We study the efficacy and mechanism of mRNA delivery using TULEP, including analysis of the cellular association, endocytosis mechanisms, endosomal escape, and protein expression in a lung cell line. We then evaluate TULEP under hypoxic conditions and address hypoxia-related deficits in efficacy by making our system tunable with adenosine triphosphate (ATP). Finally, we conclude our study with an in vivo analysis of mRNA expression, biodistribution, and tolerability of the TULEP platform in mice. In presenting these data, we hope that our work highlights the utility of TULEPs for tunable and effective mRNA delivery while more broadly highlighting the utility of considering oxygen levels when developing mRNA delivery platforms.
Collapse
Affiliation(s)
- Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
32
|
Zhang F, Burghardt T, Höhn M, Wagner E. Dual Effect by Chemical Electron Transfer Enhanced siRNA Lipid Nanoparticles: Reactive Oxygen Species-Triggered Tumor Cell Killing Aggravated by Nrf2 Gene Silencing. Pharmaceutics 2024; 16:779. [PMID: 38931900 PMCID: PMC11207527 DOI: 10.3390/pharmaceutics16060779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Insufficient endosomal escape presents a major hurdle for successful nucleic acid therapy. Here, for the first time, a chemical electron transfer (CET) system was integrated into small interfering RNA (siRNA) lipid nanoparticles (LNPs). The CET acceptor can be chemically excited using the generated energy between the donor and hydrogen peroxide, which triggers the generation of reactive oxygen species (ROS), promoting endosomal lipid membrane destabilization. Tetra-oleoyl tri-lysino succinoyl tetraethylene pentamine was included as an ionizable lipopeptide with a U-shaped topology for effective siRNA encapsulation and pH-induced endosomal escape. LNPs loaded with siRNA and CET components demonstrated a more efficient endosomal escape, as evidenced by a galectin-8-mRuby reporter; ROS significantly augmented galectin-8 recruitment by at least threefold compared with the control groups, with a p value of 0.03. Moreover, CET-enhanced LNPs achieved a 24% improvement in apoptosis level by knocking down the tumor-protective gene nuclear factor erythroid 2-related factor 2, boosting the CET-mediated ROS cell killing.
Collapse
Affiliation(s)
- Fengrong Zhang
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (T.B.); (M.H.)
| | - Tobias Burghardt
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (T.B.); (M.H.)
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (T.B.); (M.H.)
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (T.B.); (M.H.)
- CNATM-Cluster for Nucleic Acid Therapeutics Munich, 81377 Munich, Germany
- Center for Nanoscience (CeNS), LMU Munich, 81377 Munich, Germany
| |
Collapse
|
33
|
Zhang Y, Gao Z, Yang X, Xu Q, Lu Y. Leveraging high-throughput screening technologies in targeted mRNA delivery. Mater Today Bio 2024; 26:101101. [PMID: 38883419 PMCID: PMC11176929 DOI: 10.1016/j.mtbio.2024.101101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/06/2024] [Accepted: 05/25/2024] [Indexed: 06/18/2024] Open
Abstract
Messenger ribonucleic acid (mRNA) has emerged as a promising molecular preventive and therapeutic approach that opens new avenues for healthcare. Although the use of delivery systems, especially lipid nanoparticles (LNPs), greatly improves the efficiency and stability of mRNA, mRNA tends to accumulate in the liver and hardly penetrates physiological barriers to reach the target site after intravenous injection. Hence, the rational design of targeting strategies aimed at directing mRNA to specific tissues and cells remains an enormous challenge in mRNA therapy. High-throughput screening (HTS) is a cutting-edge targeted technique capable of synthesizing chemical compound libraries for the large-scale experiments to validate the efficiency of mRNA delivery system. In this review, we firstly provide an overview of conventional low-throughput targeting strategies. Then the latest advancements in HTS techniques for mRNA targeted delivery, encompassing optimizing structures of large-scale delivery vehicles and developing large-scale surface ligands, as well as the applications of HTS techniques in extrahepatic systemic diseases are comprehensively summarized. Moreover, we illustrate the selection of administration routes for targeted mRNA delivery. Finally, challenges in the field and potential solutions to tackle them are proposed, offering insights for future development toward mRNA targeted therapy.
Collapse
Affiliation(s)
- Yuchen Zhang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Zhifei Gao
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Xiao Yang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Qinglong Xu
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Yao Lu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| |
Collapse
|
34
|
Anderson CF, Singh A, Stephens T, Hoang CD, Schneider JP. Kinetically Controlled Polyelectrolyte Complex Assembly of microRNA-Peptide Nanoparticles toward Treating Mesothelioma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314367. [PMID: 38532642 PMCID: PMC11176031 DOI: 10.1002/adma.202314367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Broad size distributions and poor long-term colloidal stability of microRNA-carrying nanoparticles, especially those formed by polyelectrolyte complexation, represent major hurdles in realizing their clinical translation. Herein, peptide design is used alongside optimized flash nanocomplexation (FNC) to produce uniform peptide-based miRNA particles of exceptional stability that display anticancer activity against mesothelioma in vitro and in vivo. Modulating the content and display of lysine-based charge from small intrinsically disordered peptides used to complex miRNA proves essential in achieving stable colloids. FNC facilitates kinetic isolation of the mechanistic steps involved in particle formation to allow the preparation of particles of discrete size in a highly reproducible, scalable, and continuous manner, facilitating pre-clinical studies. To the best of the authors knowledge, this work represents the first example of employing FNC to prepare polyelectrolyte complexes of miRNA and peptide. Encapsulation of these particles into an injectable hydrogel matrix allows for their localized in vivo delivery by syringe. A one-time injection of a gel containing particles composed of miRNA-215-5p and the peptide PKM1 limits tumor progression in a xenograft model of mesothelioma.
Collapse
Affiliation(s)
- Caleb F. Anderson
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| | - Anand Singh
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tyler Stephens
- Vaccine Research Center Electron Microscopy Unit, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 20701, USA
| | - Chuong D. Hoang
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joel P. Schneider
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| |
Collapse
|
35
|
Ma Y, Tiwade PB, VanKeulen-Miller R, Narasipura EA, Fenton OS. Polyphenolic Nanoparticle Platforms (PARCELs) for In Vitro and In Vivo mRNA Delivery. NANO LETTERS 2024; 24:6092-6101. [PMID: 38728297 PMCID: PMC11218425 DOI: 10.1021/acs.nanolett.4c01235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
Despite their successful implementation in the COVID-19 vaccines, lipid nanoparticles (LNPs) still face a central limitation in the delivery of mRNA payloads: endosomal trapping. Improving upon this inefficiency could afford improved drug delivery systems, paving the way toward safer and more effective mRNA-based medicines. Here, we present polyphenolic nanoparticle platforms (PARCELs) as effective mRNA delivery systems. In brief, our investigation begins with a computationally guided structural analysis of 1825 discrete polyphenolic structural data points across 73 diverse small molecule polyphenols and 25 molecular parameters. We then generate structurally diverse PARCELs, evaluating their in vitro mechanism and activity, ultimately highlighting the superior endosomal escape properties of PARCELs relative to analogous LNPs. Finally, we examine the in vivo biodistribution, protein expression, and therapeutic efficacy of PARCELs in mice. In undertaking this approach, the goal of this study is to establish PARCELs as viable delivery platforms for safe and effective mRNA delivery.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Eshan Amruth Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen Shea Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
36
|
Yi Y, An HW, Wang H. Intelligent Biomaterialomics: Molecular Design, Manufacturing, and Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305099. [PMID: 37490938 DOI: 10.1002/adma.202305099] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/14/2023] [Indexed: 07/27/2023]
Abstract
Materialomics integrates experiment, theory, and computation in a high-throughput manner, and has changed the paradigm for the research and development of new functional materials. Recently, with the rapid development of high-throughput characterization and machine-learning technologies, the establishment of biomaterialomics that tackles complex physiological behaviors has become accessible. Breakthroughs in the clinical translation of nanoparticle-based therapeutics and vaccines have been observed. Herein, recent advances in biomaterials, including polymers, lipid-like materials, and peptides/proteins, discovered through high-throughput screening or machine learning-assisted methods, are summarized. The molecular design of structure-diversified libraries; high-throughput characterization, screening, and preparation; and, their applications in drug delivery and clinical translation are discussed in detail. Furthermore, the prospects and main challenges in future biomaterialomics and high-throughput screening development are highlighted.
Collapse
Affiliation(s)
- Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
37
|
Shen G, Liu J, Yang H, Xie N, Yang Y. mRNA therapies: Pioneering a new era in rare genetic disease treatment. J Control Release 2024; 369:696-721. [PMID: 38580137 DOI: 10.1016/j.jconrel.2024.03.056] [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: 11/04/2023] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024]
Abstract
Rare genetic diseases, often referred to as orphan diseases due to their low prevalence and limited treatment options, have long posed significant challenges to our medical system. In recent years, Messenger RNA (mRNA) therapy has emerged as a highly promising treatment approach for various diseases caused by genetic mutations. Chemically modified mRNA is introduced into cells using carriers like lipid-based nanoparticles (LNPs), producing functional proteins that compensate for genetic deficiencies. Given the advantages of precise dosing, biocompatibility, transient expression, and minimal risk of genomic integration, mRNA therapies can safely and effectively correct genetic defects in rare diseases and improve symptoms. Currently, dozens of mRNA drugs targeting rare diseases are undergoing clinical trials. This comprehensive review summarizes the progress of mRNA therapy in treating rare genetic diseases. It introduces the development, molecular design, and delivery systems of mRNA therapy, highlighting their research progress in rare genetic diseases based on protein replacement and gene editing. The review also summarizes research progress in various rare disease models and clinical trials. Additionally, it discusses the challenges and future prospects of mRNA therapy. Researchers are encouraged to join this field and collaborate to advance the clinical translation of mRNA therapy, bringing hope to patients with rare genetic diseases.
Collapse
Affiliation(s)
- Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Yang Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
38
|
Lin S, Jing H, Du X, Yang X, Wang J. Optimization of lipid assisted polymeric nanoparticles for siRNA delivery and cancer immunotherapy. Biomater Sci 2024; 12:2057-2066. [PMID: 38469870 DOI: 10.1039/d3bm02071a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
To date, five siRNA-based medications have received clinical approval and have demonstrated remarkable therapeutic efficacy in treating various diseases. However, their application has been predominantly limited to liver-specific diseases due to constraints in siRNA delivery capabilities. In this study, we have developed a siRNA delivery system utilizing clinically approved mPEG-b-PLGA, a cationic lipid, and an ionizable lipid. We optimized this system by carefully adjusting their mass ratios, resulting in highly efficient gene silencing. Furthermore, the optimized nanoparticle formulation, which encapsulates siRNA targeting CD47, induces a robust immune response. This response effectively suppresses the progression of melanoma tumors by blocking this critical immune checkpoint.
Collapse
Affiliation(s)
- Song Lin
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China.
| | - Houjin Jing
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China.
| | - Xiaojiao Du
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China.
| | - Xianzhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China.
| |
Collapse
|
39
|
Waheed I, Ali A, Tabassum H, Khatoon N, Lai WF, Zhou X. Lipid-based nanoparticles as drug delivery carriers for cancer therapy. Front Oncol 2024; 14:1296091. [PMID: 38660132 PMCID: PMC11040677 DOI: 10.3389/fonc.2024.1296091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/22/2024] [Indexed: 04/26/2024] Open
Abstract
Cancer is a severe disease that results in death in all countries of the world. A nano-based drug delivery approach is the best alternative, directly targeting cancer tumor cells with improved drug cellular uptake. Different types of nanoparticle-based drug carriers are advanced for the treatment of cancer, and to increase the therapeutic effectiveness and safety of cancer therapy, many substances have been looked into as drug carriers. Lipid-based nanoparticles (LBNPs) have significantly attracted interest recently. These natural biomolecules that alternate to other polymers are frequently recycled in medicine due to their amphipathic properties. Lipid nanoparticles typically provide a variety of benefits, including biocompatibility and biodegradability. This review covers different classes of LBNPs, including their characterization and different synthesis technologies. This review discusses the most significant advancements in lipid nanoparticle technology and their use in medicine administration. Moreover, the review also emphasized the applications of lipid nanoparticles that are used in different cancer treatment types.
Collapse
Affiliation(s)
- Ibtesam Waheed
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Anwar Ali
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Department of Biochemical and Biotechnological Sciences, School of Precision Medicine, University of Campania, Naples, Italy
| | - Huma Tabassum
- Institute of Social and Cultural Studies, Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Narjis Khatoon
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Wing-Fu Lai
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- School of Food Science and Nutrition, University of Leeds, Leeds, United Kingdom
| | - Xin Zhou
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
40
|
Mehta MJ, Kim HJ, Lim SB, Naito M, Miyata K. Recent Progress in the Endosomal Escape Mechanism and Chemical Structures of Polycations for Nucleic Acid Delivery. Macromol Biosci 2024; 24:e2300366. [PMID: 38226723 DOI: 10.1002/mabi.202300366] [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: 08/10/2023] [Revised: 12/22/2023] [Indexed: 01/17/2024]
Abstract
Nucleic acid-based therapies are seeing a spiralling surge. Stimuli-responsive polymers, especially pH-responsive ones, are gaining widespread attention because of their ability to efficiently deliver nucleic acids. These polymers can be synthesized and modified according to target requirements, such as delivery sites and the nature of nucleic acids. In this regard, the endosomal escape mechanism of polymer-nucleic acid complexes (polyplexes) remains a topic of considerable interest owing to various plausible escape mechanisms. This review describes current progress in the endosomal escape mechanism of polyplexes and state-of-the-art chemical designs for pH-responsive polymers. The importance is also discussed of the acid dissociation constant (i.e., pKa) in designing the new generation of pH-responsive polymers, along with assays to monitor and quantify the endosomal escape behavior. Further, the use of machine learning is addressed in pKa prediction and polymer design to find novel chemical structures for pH responsiveness. This review will facilitate the design of new pH-responsive polymers for advanced and efficient nucleic acid delivery.
Collapse
Affiliation(s)
- Mohit J Mehta
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Hyun Jin Kim
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
- Department of Biological Engineering, College of Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Sung Been Lim
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Mitsuru Naito
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
41
|
Huang X, Li X, Tay A. Advances in techniques to characterize cell-nanomaterial interactions (CNI). NANO TODAY 2024; 55:102149. [DOI: 10.1016/j.nantod.2024.102149] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Roh EH, Sullivan MO, Epps TH. Which Lipid Nanoparticle (LNP) Designs Work? A Simple Kinetic Model Linking LNP Chemical Structure to In Vivo Delivery Performance. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13399-13410. [PMID: 38466900 DOI: 10.1021/acsami.3c15424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Although lipid nanoparticles (LNPs) are the predominant nanocarriers for short-interfering RNA (siRNA) delivery, most therapies use nearly identical formulations that have taken 30 years to design but lack the diverse property ranges necessary for versatile application. This dearth in variety and the extended timeline for implementation are attributed to a limited understanding of how LNP properties facilitate overcoming biological barriers. Herein, a simple kinetic model was developed by using major rate-limiting steps for siRNA delivery, and this model enabled the identification of a critical parameter to predict LNP efficacy without extensive experimental testing. A volume-averaged log D, the "solubility" of charged molecules as a function of pH weighted by component volume fractions, resulted in a good correlation between LNP composition and siRNA delivery. Both the effects of modifying the structures of ionizable lipids and LNP composition on gene silencing were easily captured in the model predictions. Thus, this approach provides a robust LNP structure-activity relationship to dramatically accelerate the realization of effective LNP formulations.
Collapse
Affiliation(s)
- Esther H Roh
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Thomas H Epps
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
- Center for Research in Soft matter and Polymers (CRiSP), University of Delaware, Newark Delaware 19716, United States
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
43
|
Le ND, Nguyen BL, Patil BR, Chun H, Kim S, Nguyen TOO, Mishra S, Tandukar S, Chang JH, Kim DY, Jin SG, Choi HG, Ku SK, Kim J, Kim JO. Antiangiogenic Therapeutic mRNA Delivery Using Lung-Selective Polymeric Nanomedicine for Lung Cancer Treatment. ACS NANO 2024; 18:8392-8410. [PMID: 38450656 DOI: 10.1021/acsnano.3c13039] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Therapeutic antibodies that block vascular endothelial growth factor (VEGF) show clinical benefits in treating nonsmall cell lung cancers (NSCLCs) by inhibiting tumor angiogenesis. Nonetheless, the therapeutic effects of systemically administered anti-VEGF antibodies are often hindered in NSCLCs because of their limited distribution in the lungs and their adverse effects on normal tissues. These challenges can be overcome by delivering therapeutic antibodies in their mRNA form to lung endothelial cells, a primary target of VEGF-mediated pulmonary angiogenesis, to suppress the NSCLCs. In this study, we synthesized derivatives of poly(β-amino esters) (PBAEs) and prepared nanoparticles to encapsulate the synthetic mRNA encoding bevacizumab, an anti-VEGF antibody used in the clinic. Optimization of nanoparticle formulations resulted in a selective lung transfection after intravenous administration. Notably, the optimized PBAE nanoparticles were distributed in lung endothelial cells, resulting in the secretion of bevacizumab. We analyzed the protein corona on the lung- and spleen-targeting nanoparticles using proteomics and found distinctive features potentially contributing to their organ-selectivity. Lastly, bevacizumab mRNA delivered by the lung-targeting PBAE nanoparticles more significantly inhibited tumor proliferation and angiogenesis than recombinant bevacizumab protein in orthotopic NSCLC mouse models, supporting the therapeutic potential of bevacizumab mRNA therapy and its selective delivery through lung-targeting nanoparticles. Our proof-of-principle results highlight the clinical benefits of nanoparticle-mediated mRNA therapy in anticancer antibody treatment in preclinical models.
Collapse
Affiliation(s)
- Ngoc Duy Le
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Bao Loc Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | | | - HeeSang Chun
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - SiYoon Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | | | - Sunil Mishra
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Sudarshan Tandukar
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Jae-Hoon Chang
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Dong Young Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, Cheonan, 31116, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, Ansan, 15588, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Jeonghwan Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| |
Collapse
|
44
|
Hu Y, Eder BA, Lin J, Li S, Zhu Y, Wang TH, Guo T, Mao HQ. Liter-scale manufacturing of shelf-stable plasmid DNA/PEI transfection particles for viral vector production. Mol Ther Methods Clin Dev 2024; 32:101194. [PMID: 38352269 PMCID: PMC10863326 DOI: 10.1016/j.omtm.2024.101194] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
The transfection efficiency and stability of the delivery vehicles of plasmid DNA (pDNA) are critical metrics to ensure high-quality and high-yield production of viral vectors. We previously identified that the optimal size of pDNA/poly(ethylenimine) (PEI) transfection particles is 400-500 nm and developed a bottom-up assembly method to construct stable 400-nm pDNA/PEI particles and benchmarked their transfection efficiency in producing lentiviral vectors (LVVs). Here, we report scale-up production protocols for such transfection particles. Using a two-inlet confined impinging jet (CIJ) mixer with a dual syringe pump set-up, we produced a 1-L batch at a flow rate of 100 mL/min, and further scaled up this process with a larger CIJ mixer and a dual peristaltic pump array, allowing for continuous production at a flow rate of 1 L/min without a lot size limit. We demonstrated the scalability of this process with a 5-L lot and validated the quality of these 400-nm transfection particles against the target product profile, including physical properties, shelf and on-bench stability, transfection efficiency, and LVV production yield in both 15-mL bench culture and 2-L bioreactor runs. These results confirm the potential of this particle assembly process as a scalable manufacturing platform for viral vector production.
Collapse
Affiliation(s)
- Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | - Jinghan Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sixuan Li
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Tza-Huei Wang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ting Guo
- 2seventy bio, Inc., Cambridge, MA 02142, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
45
|
Witten J, Hu Y, Langer R, Anderson DG. Recent advances in nanoparticulate RNA delivery systems. Proc Natl Acad Sci U S A 2024; 121:e2307798120. [PMID: 38437569 PMCID: PMC10945842 DOI: 10.1073/pnas.2307798120] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Nanoparticle-based RNA delivery has shown great progress in recent years with the approval of two mRNA vaccines for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and a liver-targeted siRNA therapy. Here, we discuss the preclinical and clinical advancement of new generations of RNA delivery therapies along multiple axes. Improvements in cargo design such as RNA circularization and data-driven untranslated region optimization can drive better mRNA expression. New materials discovery research has driven improved delivery to extrahepatic targets such as the lung and splenic immune cells, which could lead to pulmonary gene therapy and better cancer vaccines, respectively. Other organs and even specific cell types can be targeted for delivery via conjugation of small molecule ligands, antibodies, or peptides to RNA delivery nanoparticles. Moreover, the immune response to any RNA delivery nanoparticle plays a crucial role in determining efficacy. Targeting increased immunogenicity without induction of reactogenic side effects is crucial for vaccines, while minimization of immune response is important for gene therapies. New developments have addressed each of these priorities. Last, we discuss the range of RNA delivery clinical trials targeting diverse organs, cell types, and diseases and suggest some key advances that may play a role in the next wave of therapies.
Collapse
Affiliation(s)
- Jacob Witten
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Yizong Hu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard and Massachusetts Institute of Technology Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA02115
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Daniel G. Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard and Massachusetts Institute of Technology Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA02115
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
46
|
Chatterjee S, Kon E, Sharma P, Peer D. Endosomal escape: A bottleneck for LNP-mediated therapeutics. Proc Natl Acad Sci U S A 2024; 121:e2307800120. [PMID: 38437552 PMCID: PMC10945858 DOI: 10.1073/pnas.2307800120] [Citation(s) in RCA: 97] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Lipid nanoparticles (LNPs) have recently emerged as a powerful and versatile clinically approved platform for nucleic acid delivery, specifically for mRNA vaccines. A major bottleneck in the field is the release of mRNA-LNPs from the endosomal pathways into the cytosol of cells where they can execute their encoded functions. The data regarding the mechanism of these endosomal escape processes are limited and contradicting. Despite extensive research, there is no consensus regarding the compartment of escape, the cause of the inefficient escape and are currently lacking a robust method to detect the escape. Here, we review the currently known mechanisms of endosomal escape and the available methods to study this process. We critically discuss the limitations and challenges of these methods and the possibilities to overcome these challenges. We propose that the development of currently lacking robust, quantitative high-throughput techniques to study endosomal escape is timely and essential. A better understanding of this process will enable better RNA-LNP designs with improved efficiency to unlock new therapeutic modalities.
Collapse
Affiliation(s)
- Sushmita Chatterjee
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Edo Kon
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Preeti Sharma
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
47
|
Shen J, Lima e Silva R, Zhang M, Luly KM, Hackett SF, Tzeng SY, Lowmaster SM, Shannon SR, Wilson DR, Green JJ, Campochiaro PA. Suprachoroidal gene transfer with nonviral nanoparticles in large animal eyes. SCIENCE ADVANCES 2024; 10:eadl3576. [PMID: 38457512 PMCID: PMC10923522 DOI: 10.1126/sciadv.adl3576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/01/2024] [Indexed: 03/10/2024]
Abstract
Suprachoroidal nonviral gene therapy with biodegradable poly(β-amino ester) nanoparticles (NPs) provides widespread expression in photoreceptors and retinal pigmented epithelial (RPE) cells and therapeutic benefits in rodents. Here, we show in a human-sized minipig eye that suprachoroidal injection of 50 μl of NPs containing 19.2 μg of GFP expression plasmid caused GFP expression in photoreceptors and RPE throughout the entire eye with no toxicity. Two weeks after injection of 50, 100, or 200 μl, there was considerable within-eye and between-eye variability in expression that was reduced 3 months after injection of 200 μl and markedly reduced after three suprachoroidal injections at different locations around the eye. Reduction of bacterial CpG sequences in the expression plasmid resulted in a trend toward higher expression. These data indicate that nonviral suprachoroidal gene therapy with optimized polymer, expression plasmid, and injection approach has potential for treating photoreceptors throughout the entire retina of a human-sized eye.
Collapse
Affiliation(s)
- Jikui Shen
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raquel Lima e Silva
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mingliang Zhang
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn M. Luly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sean F. Hackett
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shirley M. Lowmaster
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sydney R. Shannon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David R. Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jordan J. Green
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A. Campochiaro
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
48
|
Xue L, Hamilton AG, Zhao G, Xiao Z, El-Mayta R, Han X, Gong N, Xiong X, Xu J, Figueroa-Espada CG, Shepherd SJ, Mukalel AJ, Alameh MG, Cui J, Wang K, Vaughan AE, Weissman D, Mitchell MJ. High-throughput barcoding of nanoparticles identifies cationic, degradable lipid-like materials for mRNA delivery to the lungs in female preclinical models. Nat Commun 2024; 15:1884. [PMID: 38424061 PMCID: PMC10904786 DOI: 10.1038/s41467-024-45422-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Lipid nanoparticles for delivering mRNA therapeutics hold immense promise for the treatment of a wide range of lung-associated diseases. However, the lack of effective methodologies capable of identifying the pulmonary delivery profile of chemically distinct lipid libraries poses a significant obstacle to the advancement of mRNA therapeutics. Here we report the implementation of a barcoded high-throughput screening system as a means to identify the lung-targeting efficacy of cationic, degradable lipid-like materials. We combinatorially synthesize 180 cationic, degradable lipids which are initially screened in vitro. We then use barcoding technology to quantify how the selected 96 distinct lipid nanoparticles deliver DNA barcodes in vivo. The top-performing nanoparticle formulation delivering Cas9-based genetic editors exhibits therapeutic potential for antiangiogenic cancer therapy within a lung tumor model in female mice. These data demonstrate that employing high-throughput barcoding technology as a screening tool for identifying nanoparticles with lung tropism holds potential for the development of next-generation extrahepatic delivery platforms.
Collapse
Affiliation(s)
- Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alex G Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gan Zhao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zebin Xiao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xinhong Xiong
- Yangtze Delta Region Institute (Huzhou), University of Electronic Science and Technology of China, Huzhou, Zhejiang, 313001, China
| | - Junchao Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Sarah J Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alvin J Mukalel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jiaxi Cui
- Yangtze Delta Region Institute (Huzhou), University of Electronic Science and Technology of China, Huzhou, Zhejiang, 313001, China
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, 19122, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19014, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
49
|
Reshma G B, Miglani C, Pal A, Ganguli M. Sugar alcohol-modified polyester nanoparticles for gene delivery via selective caveolae-mediated endocytosis. NANOSCALE 2024; 16:4114-4124. [PMID: 38353098 DOI: 10.1039/d3nr05300h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Nucleic acid-based drugs are changing the scope of emerging medicine in preventing and treating diseases. Nanoparticle systems based on lipids and polymers developed to navigate tissue-level and cellular-level barriers are now emerging as vector systems that can be translated to clinical settings. A class of polymers, poly(β-amino esters) (PBAEs) known for their chemical flexibility and biodegradability, has been explored for gene delivery. These polymers are sensitive to changes in the monomer composition affecting transfection efficiency. Hence to add functionality to these polymers, we partially substituted ligands to an identified effective polymer chemistry. We report here a new series of statistical copolymers based on PBAEs where the backbone is modified with sugar alcohols to selectively facilitate the caveolae-mediated endocytosis pathway of cellular transport. These ligands are grafted at the polymer's backbone, thereby establishing a new strategy of modification in PBAEs. We demonstrate that these polymers form nanoparticles with DNA, show effective complexation and cargo release, enter the cell via selective caveolae-mediated endocytosis, exhibit low cytotoxicity, and increase transfection in neuronal cells.
Collapse
Affiliation(s)
- Betsy Reshma G
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chirag Miglani
- Chemical Biology Unit, Institute of Nanoscience and Technology, Sector 81, Mohali, Punjab 140306, India
| | - Asish Pal
- Chemical Biology Unit, Institute of Nanoscience and Technology, Sector 81, Mohali, Punjab 140306, India
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110025, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
50
|
Hamilton S, Kingston BR. Applying artificial intelligence and computational modeling to nanomedicine. Curr Opin Biotechnol 2024; 85:103043. [PMID: 38091874 DOI: 10.1016/j.copbio.2023.103043] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 02/09/2024]
Abstract
Achieving specific and targeted delivery of nanomedicines to diseased tissues is a major challenge. This is because the process of designing, formulating, testing, and selecting a nanoparticle delivery vehicle for a specific disease target is governed by complex multivariate interactions. Computational modeling and artificial intelligence are well-suited for analyzing and modeling large multivariate datasets in short periods of time. Computational approaches can be applied to help design nanomedicine formulations, interpret nanoparticle-biological interactions, and create models from high-throughput screening techniques to improve the selection of the ideal nanoparticle carrier. In the future, many steps in the nanomedicine development process will be done computationally, reducing the number of experiments and time needed to select the ideal nanomedicine formulation.
Collapse
Affiliation(s)
- Sean Hamilton
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, 2720 S. Moody Avenue, Portland, OR 97201, United States
| | - Benjamin R Kingston
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, 2720 S. Moody Avenue, Portland, OR 97201, United States.
| |
Collapse
|