1
|
Chernyi N, Gavrilova D, Saruhanyan M, Oloruntimehin ES, Karabelsky A, Bezsonov E, Malogolovkin A. Recent Advances in Gene Therapy for Hemophilia: Projecting the Perspectives. Biomolecules 2024; 14:854. [PMID: 39062568 PMCID: PMC11274510 DOI: 10.3390/biom14070854] [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: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
One of the well-known X-linked genetic disorders is hemophilia, which could be hemophilia A as a result of a mutation in the F8 (factor VIII) gene or hemophilia B as a result of a mutation in the F9 (factor IX) gene, leading to insufficient levels of the proteins essential for blood coagulation cascade. In patients with severe hemophilia, factor VIII or factor IX activities in the blood plasma are considerably low, estimated to be less than 1%. This is responsible for spontaneous or post-traumatic bleeding episodes, or both, leading to disease complications and death. Current treatment of hemophilia relies on the prevention of bleeding, which consists of expensive lifelong replacement infusion therapy of blood plasma clotting factors, their recombinant versions, or therapy with recombinant monoclonal antibodies. Recently emerged gene therapy approaches may be a potential game changer that could reshape the therapeutic outcomes of hemophilia A or B using a one-off vector in vivo delivery and aim to achieve long-term endogenous expression of factor VIII or IX. This review examines both traditional approaches to the treatment of hemophilia and modern methods, primarily focusing on gene therapy, to update knowledge in this area. Recent technological advances and gene therapeutics in the pipeline are critically reviewed and summarized. We consider gene therapy to be the most promising method as it may overcome the problems associated with more traditional treatments, such as the need for constant and expensive infusions and the presence of an immune response to the antibody drugs used to treat hemophilia.
Collapse
Affiliation(s)
- Nikita Chernyi
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Darina Gavrilova
- Department of Biology and General Genetics, First Moscow State Medical University (Sechenov University), Moscow 105043, Russia
| | - Mane Saruhanyan
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Ezekiel S. Oloruntimehin
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Alexander Karabelsky
- Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354530, Russia
| | - Evgeny Bezsonov
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Department of Biology and General Genetics, First Moscow State Medical University (Sechenov University), Moscow 105043, Russia
| | - Alexander Malogolovkin
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Center for Translational Medicine, Sirius University of Science and Technology, Sochi 354530, Russia
| |
Collapse
|
2
|
Zhang W, Hou Y, Yin S, Miao Q, Lee K, Zhou X, Wang Y. Advanced gene nanocarriers/scaffolds in nonviral-mediated delivery system for tissue regeneration and repair. J Nanobiotechnology 2024; 22:376. [PMID: 38926780 PMCID: PMC11200991 DOI: 10.1186/s12951-024-02580-8] [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: 03/09/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Tissue regeneration technology has been rapidly developed and widely applied in tissue engineering and repair. Compared with traditional approaches like surgical treatment, the rising gene therapy is able to have a durable effect on tissue regeneration, such as impaired bone regeneration, articular cartilage repair and cancer-resected tissue repair. Gene therapy can also facilitate the production of in situ therapeutic factors, thus minimizing the diffusion or loss of gene complexes and enabling spatiotemporally controlled release of gene products for tissue regeneration. Among different gene delivery vectors and supportive gene-activated matrices, advanced gene/drug nanocarriers attract exceptional attraction due to their tunable physiochemical properties, as well as excellent adaptive performance in gene therapy for tissue regeneration, such as bone, cartilage, blood vessel, nerve and cancer-resected tissue repair. This paper reviews the recent advances on nonviral-mediated gene delivery systems with an emphasis on the important role of advanced nanocarriers in gene therapy and tissue regeneration.
Collapse
Affiliation(s)
- Wanheng Zhang
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yan Hou
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, 200444, China
| | - Shiyi Yin
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi Miao
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kyubae Lee
- Department of Biomedical Materials, Konyang University, Daejeon, 35365, Republic of Korea
| | - Xiaojian Zhou
- Department of Pediatrics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Yongtao Wang
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Shanghai, 200444, China.
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
3
|
Petrovic S, Bita B, Barbinta-Patrascu ME. Nanoformulations in Pharmaceutical and Biomedical Applications: Green Perspectives. Int J Mol Sci 2024; 25:5842. [PMID: 38892030 PMCID: PMC11172476 DOI: 10.3390/ijms25115842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
This study provides a brief discussion of the major nanopharmaceuticals formulations as well as the impact of nanotechnology on the future of pharmaceuticals. Effective and eco-friendly strategies of biofabrication are also highlighted. Modern approaches to designing pharmaceutical nanoformulations (e.g., 3D printing, Phyto-Nanotechnology, Biomimetics/Bioinspiration, etc.) are outlined. This paper discusses the need to use natural resources for the "green" design of new nanoformulations with therapeutic efficiency. Nanopharmaceuticals research is still in its early stages, and the preparation of nanomaterials must be carefully considered. Therefore, safety and long-term effects of pharmaceutical nanoformulations must not be overlooked. The testing of nanopharmaceuticals represents an essential point in their further applications. Vegetal scaffolds obtained by decellularizing plant leaves represent a valuable, bioinspired model for nanopharmaceutical testing that avoids using animals. Nanoformulations are critical in various fields, especially in pharmacy, medicine, agriculture, and material science, due to their unique properties and advantages over conventional formulations that allows improved solubility, bioavailability, targeted drug delivery, controlled release, and reduced toxicity. Nanopharmaceuticals have transitioned from experimental stages to being a vital component of clinical practice, significantly improving outcomes in medical fields for cancer treatment, infectious diseases, neurological disorders, personalized medicine, and advanced diagnostics. Here are the key points highlighting their importance. The significant challenges, opportunities, and future directions are mentioned in the final section.
Collapse
Affiliation(s)
- Sanja Petrovic
- Department of Chemical Technologies, Faculty of Technology, University of Nis, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia;
| | - Bogdan Bita
- Department of Electricity, Solid-State Physics and Biophysics, Faculty of Physics, University of Bucharest, 405 Atomistilor Street, P.O. Box MG-11, 077125 Magurele, Romania;
| | - Marcela-Elisabeta Barbinta-Patrascu
- Department of Electricity, Solid-State Physics and Biophysics, Faculty of Physics, University of Bucharest, 405 Atomistilor Street, P.O. Box MG-11, 077125 Magurele, Romania;
| |
Collapse
|
4
|
Ma X, Zhang Y, Huang K, Zhu L, Xu W. Multifunctional rolling circle transcription-based nanomaterials for advanced drug delivery. Biomaterials 2023; 301:122241. [PMID: 37451000 DOI: 10.1016/j.biomaterials.2023.122241] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/21/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
As the up-and-comer in the development of RNA nanotechnology, RNA nanomaterials based on functionalized rolling circle transcription (RCT) have become promising carriers for drug production and delivery. This is due to RCT technology can self-produce polyvalent tandem nucleic acid prodrugs for intervention in intracellular gene expression and protein production. RNA component strands participating in de novo assembly enable RCT-based nanomaterials to exhibit good mechanical properties, biostability, and biocompatibility as delivery carriers. The biostability makes it to suitable for thermodynamically/kinetically favorable assembly, enzyme resistance and efficient expression in vivo. Controllable RCT system combined with polymers enables customizable and adjustable size, shape, structure, and stoichiometry of RNA building materials, which provide groundwork for the delivery of advanced drugs. Here, we review the assembly strategies and the dynamic regulation of RCT-based nanomaterials, summarize its functional properties referring to the bottom-up design philosophy, and describe its advancements in tumor gene therapy, synergistic chemotherapy, and immunotherapy. Last, we elaborate on the unique and practical value of RCT-based nanomaterials, namely "self-production and self-sale", and their potential challenges in nanotechnology, material science and biomedicine.
Collapse
Affiliation(s)
- Xuan Ma
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China; College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, 100083, China
| | - Yangzi Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China; College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, 100083, China
| | - Kunlun Huang
- College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, 100083, China
| | - Longjiao Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Wentao Xu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China; College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
5
|
Du X, Yada E, Terai Y, Takahashi T, Nakanishi H, Tanaka H, Akita H, Itaka K. Comprehensive Evaluation of Lipid Nanoparticles and Polyplex Nanomicelles for Muscle-Targeted mRNA Delivery. Pharmaceutics 2023; 15:2291. [PMID: 37765260 PMCID: PMC10536695 DOI: 10.3390/pharmaceutics15092291] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The growing significance of messenger RNA (mRNA) therapeutics in diverse medical applications, such as cancer, infectious diseases, and genetic disorders, highlighted the need for efficient and safe delivery systems. Lipid nanoparticles (LNPs) have shown great promise for mRNA delivery, but challenges such as toxicity and immunogenicity still remain to be addressed. In this study, we aimed to compare the performance of polyplex nanomicelles, our original cationic polymer-based carrier, and LNPs in various aspects, including delivery efficiency, organ toxicity, muscle damage, immune reaction, and pain. Our results showed that nanomicelles (PEG-PAsp(DET)) and LNPs (SM-102) exhibited distinct characteristics, with the former demonstrating relatively sustained protein production and reduced inflammation, making them suitable for therapeutic purposes. On the other hand, LNPs displayed desirable properties for vaccines, such as rapid mRNA expression and potent immune response. Taken together, these results suggest the different potentials of nanomicelles and LNPs, supporting further optimization of mRNA delivery systems tailored for specific purposes.
Collapse
Affiliation(s)
- Xuan Du
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Erica Yada
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
- NANO MRNA, Co., Ltd. Tokyo 104-0031, Japan
| | - Yuki Terai
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Takuya Takahashi
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Hideyuki Nakanishi
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Hiroki Tanaka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Hidetaka Akita
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
- Clinical Biotechnology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
6
|
Uchida S, Lau CYJ, Oba M, Miyata K. Polyplex designs for improving the stability and safety of RNA therapeutics. Adv Drug Deliv Rev 2023; 199:114972. [PMID: 37364611 DOI: 10.1016/j.addr.2023.114972] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Nanoparticle-based delivery systems have contributed to the recent clinical success of RNA therapeutics, including siRNA and mRNA. RNA delivery using polymers has several distinct properties, such as enabling RNA delivery into extra-hepatic organs, modulation of immune responses to RNA, and regulation of intracellular RNA release. However, delivery systems should overcome safety and stability issues to achieve widespread therapeutic applications. Safety concerns include direct damage to cellular components, innate and adaptive immune responses, complement activation, and interaction with surrounding molecules and cells in the blood circulation. The stability of the delivery systems should balance extracellular RNA protection and controlled intracellular RNA release, which requires optimization for each RNA species. Further, polymer designs for improving safety and stability often conflict with each other. This review covers advances in polymer-based approaches to address these issues over several years, focusing on biological understanding and design concepts for delivery systems rather than material chemistry.
Collapse
Affiliation(s)
- Satoshi Uchida
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto, 606-0823, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| | - Chun Yin Jerry Lau
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Makoto Oba
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto, 606-0823, 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
|
7
|
Tockary TA, Abbasi S, Matsui-Masai M, Hayashi A, Yoshinaga N, Boonstra E, Wang Z, Fukushima S, Kataoka K, Uchida S. Comb-structured mRNA vaccine tethered with short double-stranded RNA adjuvants maximizes cellular immunity for cancer treatment. Proc Natl Acad Sci U S A 2023; 120:e2214320120. [PMID: 37428918 PMCID: PMC10629565 DOI: 10.1073/pnas.2214320120] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 06/09/2023] [Indexed: 07/12/2023] Open
Abstract
Integrating antigen-encoding mRNA (Messenger RNA) and immunostimulatory adjuvant into a single formulation is a promising approach to potentiating the efficacy of mRNA vaccines. Here, we developed a scheme based on RNA engineering to integrate adjuvancy directly into antigen-encoding mRNA strands without hampering the ability to express antigen proteins. Short double-stranded RNA (dsRNA) was designed to target retinoic acid-inducible gene-I (RIG-I), an innate immune receptor, for effective cancer vaccination and then tethered onto the mRNA strand via hybridization. Tuning the dsRNA structure and microenvironment by changing its length and sequence enabled the determination of the structure of dsRNA-tethered mRNA efficiently stimulating RIG-I. Eventually, the formulation loaded with dsRNA-tethered mRNA of the optimal structure effectively activated mouse and human dendritic cells and drove them to secrete a broad spectrum of proinflammatory cytokines without increasing the secretion of anti-inflammatory cytokines. Notably, the immunostimulating intensity was tunable by modulating the number of dsRNA along the mRNA strand, which prevents excessive immunostimulation. Versatility in the applicable formulation is a practical advantage of the dsRNA-tethered mRNA. Its formulation with three existing systems, i.e., anionic lipoplex, ionizable lipid-based lipid nanoparticles, and polyplex micelles, induced appreciable cellular immunity in the mice model. Of particular interest, dsRNA-tethered mRNA encoding ovalbumin (OVA) formulated in anionic lipoplex used in clinical trials exerted a significant therapeutic effect in the mouse lymphoma (E.G7-OVA) model. In conclusion, the system developed here provides a simple and robust platform to supply the desired intensity of immunostimulation in various formulations of mRNA cancer vaccines.
Collapse
Affiliation(s)
- Theofilus A. Tockary
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki210-0821, Japan
| | - Saed Abbasi
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki210-0821, Japan
| | - Miki Matsui-Masai
- Department of Research, NanoCarrier Co., Ltd., Kawasaki-ku, Kawasaki210-0821, Japan
| | - Akimasa Hayashi
- Department of Pathology, Kyorin University School of Medicine, Mitaka-shi, Tokyo181-8611, Japan
| | - Naoto Yoshinaga
- Biomacromolecule Research Team, RIKEN Center for Sustainable Resource Science, Saitama351-0198, Japan
| | - Eger Boonstra
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo113-8656, Japan
| | - Zheng Wang
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki210-0821, Japan
| | - Shigeto Fukushima
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki210-0821, Japan
- Department of Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto606-0823, Japan
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
| |
Collapse
|
8
|
Zhang M, Fukushima Y, Nozaki K, Nakanishi H, Deng J, Wakabayashi N, Itaka K. Enhancement of bone regeneration by coadministration of angiogenic and osteogenic factors using messenger RNA. Inflamm Regen 2023; 43:32. [PMID: 37340499 DOI: 10.1186/s41232-023-00285-3] [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: 03/01/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Bone defects remain a challenge today. In addition to osteogenic activation, the crucial role of angiogenesis has also gained attention. In particular, vascular endothelial growth factor (VEGF) is likely to play a significant role in bone regeneration, not only to restore blood supply but also to be directly involved in the osteogenic differentiation of mesenchymal stem cells. In this study, to produce additive angiogenic-osteogenic effects in the process of bone regeneration, VEGF and Runt-related transcription factor 2 (Runx2), an essential transcription factor for osteogenic differentiation, were coadministered with messenger RNAs (mRNAs) to bone defects in the rat mandible. METHODS The mRNAs encoding VEGF or Runx2 were prepared via in vitro transcription (IVT). Osteogenic differentiation after mRNA transfection was evaluated using primary osteoblast-like cells, followed by an evaluation of the gene expression levels of osteogenic markers. The mRNAs were then administered to a bone defect prepared in the rat mandible using our original cationic polymer-based carrier, the polyplex nanomicelle. The bone regeneration was evaluated by micro-computerized tomography (μCT) imaging, and histologic analyses. RESULTS Osteogenic markers such as osteocalcin (Ocn) and osteopontin (Opn) were significantly upregulated after mRNA transfection. VEGF mRNA was revealed to have a distinct osteoblastic function similar to that of Runx2 mRNA, and the combined use of the two mRNAs resulted in further upregulation of the markers. After in vivo administration into the bone defect, the two mRNAs induced significant enhancement of bone regeneration with increased bone mineralization. Histological analyses using antibodies against the Cluster of Differentiation 31 protein (CD31), alkaline phosphatase (ALP), or OCN revealed that the mRNAs induced the upregulation of osteogenic markers in the defect, together with increased vessel formation, leading to rapid bone formation. CONCLUSIONS These results demonstrate the feasibility of using mRNA medicines to introduce various therapeutic factors, including transcription factors, into target sites. This study provides valuable information for the development of mRNA therapeutics for tissue engineering.
Collapse
Affiliation(s)
- Maorui Zhang
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, 1010062, Japan
- Department of Advanced Prosthodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 1138549, Japan
- Department of Oral Implantology, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Yuta Fukushima
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, 1010062, Japan
| | - Kosuke Nozaki
- Department of Advanced Prosthodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 1138549, Japan
| | - Hideyuki Nakanishi
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, 1010062, Japan
| | - Jia Deng
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, 1010062, Japan
- Department of Masticatory Function and Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8549, Japan
| | - Noriyuki Wakabayashi
- Department of Advanced Prosthodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 1138549, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, 1010062, Japan.
- Clinical Biotechnology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
9
|
Liu C, Shi Q, Huang X, Koo S, Kong N, Tao W. mRNA-based cancer therapeutics. Nat Rev Cancer 2023:10.1038/s41568-023-00586-2. [PMID: 37311817 DOI: 10.1038/s41568-023-00586-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 06/15/2023]
Abstract
Due to the fact that mRNA technology allows the production of diverse vaccines and treatments in a shorter time frame and with reduced expense compared to conventional approaches, there has been a surge in the use of mRNA-based therapeutics in recent years. With the aim of encoding tumour antigens for cancer vaccines, cytokines for immunotherapy, tumour suppressors to inhibit tumour development, chimeric antigen receptors for engineered T cell therapy or genome-editing proteins for gene therapy, many of these therapeutics have shown promising efficacy in preclinical studies, and some have even entered clinical trials. Given the evidence supporting the effectiveness and safety of clinically approved mRNA vaccines, coupled with growing interest in mRNA-based therapeutics, mRNA technology is poised to become one of the major pillars in cancer drug development. In this Review, we present in vitro transcribed mRNA-based therapeutics for cancer treatment, including the characteristics of the various types of synthetic mRNA, the packaging systems for efficient mRNA delivery, preclinical and clinical studies, current challenges and future prospects in the field. We anticipate the translation of promising mRNA-based treatments into clinical applications, to ultimately benefit patients.
Collapse
Affiliation(s)
- Chuang Liu
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Qiangqiang Shi
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Xiangang Huang
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Yang W, Mixich L, Boonstra E, Cabral H. Polymer-Based mRNA Delivery Strategies for Advanced Therapies. Adv Healthc Mater 2023; 12:e2202688. [PMID: 36785927 PMCID: PMC11469255 DOI: 10.1002/adhm.202202688] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/31/2023] [Indexed: 02/15/2023]
Abstract
Messenger RNA (mRNA)-based therapies offer great promise for the treatment of a variety of diseases. In 2020, two FDA approvals of mRNA-based vaccines have elevated mRNA vaccines to global recognition. However, the therapeutic capabilities of mRNA extend far beyond vaccines against infectious diseases. They hold potential for cancer vaccines, protein replacement therapies, gene editing therapies, and immunotherapies. For realizing such advanced therapies, it is crucial to develop effective carrier systems. Recent advances in materials science have led to the development of promising nonviral mRNA delivery systems. In comparison to other carriers like lipid nanoparticles, polymer-based delivery systems often receive less attention, despite their unique ability to carefully tune their chemical features to promote mRNA protection, their favorable pharmacokinetics, and their potential for targeting delivery. In this review, the central features of polymer-based systems for mRNA delivery highlighting the molecular design criteria, stability, and biodistribution are discussed. Finally, the role of targeting ligands for the future of RNA therapies is analyzed.
Collapse
Affiliation(s)
- Wenqian Yang
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Lucas Mixich
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Eger Boonstra
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Horacio Cabral
- Department of BioengineeringGraduate School of EngineeringThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| |
Collapse
|
11
|
Hosseinkhani H, Domb AJ, Sharifzadeh G, Nahum V. Gene Therapy for Regenerative Medicine. Pharmaceutics 2023; 15:856. [PMID: 36986717 PMCID: PMC10057434 DOI: 10.3390/pharmaceutics15030856] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The development of biological methods over the past decade has stimulated great interest in the possibility to regenerate human tissues. Advances in stem cell research, gene therapy, and tissue engineering have accelerated the technology in tissue and organ regeneration. However, despite significant progress in this area, there are still several technical issues that must be addressed, especially in the clinical use of gene therapy. The aims of gene therapy include utilising cells to produce a suitable protein, silencing over-producing proteins, and genetically modifying and repairing cell functions that may affect disease conditions. While most current gene therapy clinical trials are based on cell- and viral-mediated approaches, non-viral gene transfection agents are emerging as potentially safe and effective in the treatment of a wide variety of genetic and acquired diseases. Gene therapy based on viral vectors may induce pathogenicity and immunogenicity. Therefore, significant efforts are being invested in non-viral vectors to enhance their efficiency to a level comparable to the viral vector. Non-viral technologies consist of plasmid-based expression systems containing a gene encoding, a therapeutic protein, and synthetic gene delivery systems. One possible approach to enhance non-viral vector ability or to be an alternative to viral vectors would be to use tissue engineering technology for regenerative medicine therapy. This review provides a critical view of gene therapy with a major focus on the development of regenerative medicine technologies to control the in vivo location and function of administered genes.
Collapse
Affiliation(s)
- Hossein Hosseinkhani
- Innovation Center for Advanced Technology, Matrix, Inc., New York, NY 10019, USA
| | - Abraham J. Domb
- The Center for Nanoscience and Nanotechnology, Alex Grass Center for Drug Design and Synthesis and Cannabinoids Research, School of Pharmacy, Faculty of Medicine, Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Ghorbanali Sharifzadeh
- Department of Polymer Engineering, School of Chemical Engineering, Universiti Teknologi Malaysia, Skudai 81310, Johor, Malaysia
| | - Victoria Nahum
- The Center for Nanoscience and Nanotechnology, Alex Grass Center for Drug Design and Synthesis and Cannabinoids Research, School of Pharmacy, Faculty of Medicine, Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
12
|
Lores S, Gámez-Chiachio M, Cascallar M, Ramos-Nebot C, Hurtado P, Alijas S, López López R, Piñeiro R, Moreno-Bueno G, de la Fuente M. Effectiveness of a novel gene nanotherapy based on putrescine for cancer treatment. Biomater Sci 2023. [PMID: 36790445 DOI: 10.1039/d2bm01456d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Gene therapy has long been proposed for cancer treatment. However, the use of therapeutic nucleic acids presents several limitations such as enzymatic degradation, rapid clearance, and poor cellular uptake and efficiency. In this work we propose the use of putrescine, a precursor for higher polyamine biosynthesis for the preparation of cationic nanosystems for cancer gene therapy. We have formulated and characterized putrescine-sphingomyelin nanosystems (PSN) and studied their endocytic pathway and intracellular trafficking in cancer cells. After loading a plasmid DNA (pDNA) encoding the apoptotic Fas Ligand (FasL), we proved their therapeutic activity by measuring the cell death rate after treatment of MDA-MB-231 cells. We have also used xenografted zebrafish embryos as a first in vivo approach to demonstrate the efficacy of the proposed PSN-pDNA formulation in a more complex model. Finally, intratumoral and intraperitoneal administration to mice-bearing MDA-MB-231 xenografts resulted in a significant decrease in tumour cell growth, highlighting the potential of the developed gene therapy nanoformulation for the treatment of triple negative breast cancer.
Collapse
Affiliation(s)
- Saínza Lores
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain
| | - Manuel Gámez-Chiachio
- Translational Cancer Research Laboratory, Department of Biochemistry, Autonomous University of Madrid, School of Medicine, "Alberto Sols" Biomedical Research Institute CSIC-UAM, IdiPaz, Arturo Duperier 4, 28029, Madrid, Spain. .,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain
| | - María Cascallar
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain.,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain
| | - Carmen Ramos-Nebot
- Translational Cancer Research Laboratory, Department of Biochemistry, Autonomous University of Madrid, School of Medicine, "Alberto Sols" Biomedical Research Institute CSIC-UAM, IdiPaz, Arturo Duperier 4, 28029, Madrid, Spain. .,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain
| | - Pablo Hurtado
- Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Rafael López López
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain.,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Roberto Piñeiro
- Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Gema Moreno-Bueno
- Translational Cancer Research Laboratory, Department of Biochemistry, Autonomous University of Madrid, School of Medicine, "Alberto Sols" Biomedical Research Institute CSIC-UAM, IdiPaz, Arturo Duperier 4, 28029, Madrid, Spain. .,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,MD Anderson International Foundation, Gómez Hemans s/n, 28033 Madrid, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain.,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,DIVERSA Technologies SL, Edificio Emprendia, Universidade de Santiago de Compostela, Campus Vida s/n, 15782 Santiago de Compostela, Spain
| |
Collapse
|
13
|
Son S, Lee K. Development of mRNA Vaccines/Therapeutics and Their Delivery System. Mol Cells 2023; 46:41-47. [PMID: 36697236 PMCID: PMC9880606 DOI: 10.14348/molcells.2023.2165] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/18/2022] [Accepted: 12/18/2022] [Indexed: 01/27/2023] Open
Abstract
The rapid development of mRNA vaccines has contributed to the management of the current coronavirus disease 2019 (COVID-19) pandemic, suggesting that this technology may be used to manage future outbreaks of infectious diseases. Because the antigens targeted by mRNA vaccines can be easily altered by simply changing the sequence present in the coding region of mRNA structures, it is more appropriate to develop vaccines, especially during rapidly developing outbreaks of infectious diseases. In addition to allowing rapid development, mRNA vaccines have great potential in inducing successful antigen-specific immunity by expressing target antigens in cells and simultaneously triggering immune responses. Indeed, the two COVID-19 mRNA vaccines approved by the U.S. Food and Drug Administration have shown significant efficacy in preventing infections. The ability of mRNAs to produce target proteins that are defective in specific diseases has enabled the development of options to treat intractable diseases. Clinical applications of mRNA vaccines/therapeutics require strategies to safely deliver the RNA molecules into targeted cells. The present review summarizes current knowledge about mRNA vaccines/ therapeutics, their clinical applications, and their delivery strategies.
Collapse
Affiliation(s)
- Sora Son
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Kyuri Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
14
|
Park Y, Moses AS, Demessie AA, Singh P, Lee H, Korzun T, Taratula OR, Alani AG, Taratula O. Poly(aspartic acid)-Based Polymeric Nanoparticle for Local and Systemic mRNA Delivery. Mol Pharm 2022; 19:4696-4704. [PMID: 36409995 PMCID: PMC9826779 DOI: 10.1021/acs.molpharmaceut.2c00738] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recently, therapeutics based on mRNA (mRNA) have attracted significant interest for vaccines, cancer immunotherapy, and gene editing. However, the lack of biocompatible vehicles capable of delivering mRNA to the target tissue and efficiently expressing the encoded proteins impedes the development of mRNA-based therapies for a variety of diseases. Herein, we report mRNA-loaded polymeric nanoparticles based on diethylenetriamine-substituted poly(aspartic acid) that induce protein expression in the lungs and muscles following intravenous and intramuscular injections, respectively. Animal studies revealed that the amount of polyethylene glycol (PEG) on the nanoparticle surface affects the translation of the delivered mRNA into the encoded protein in the target tissue. After systemic administration, only mRNA-loaded nanoparticles modified with PEG at a molar ratio of 1:1 (PEG/polymer) induce protein expression in the lungs. In contrast, protein expression was detected only following intramuscular injection of mRNA-loaded nanoparticles with a PEG/polymer ratio of 10:1. These findings suggest that the PEG density on the surface of poly(aspartic acid)-based nanoparticles should be optimized for different delivery routes depending on the purpose of the mRNA treatment.
Collapse
Affiliation(s)
- Youngrong Park
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Abraham S. Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Ananiya A. Demessie
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Prem Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Hyelim Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Olena R. Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Adam G. Alani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97239, United States
| |
Collapse
|
15
|
Deng J, Fukushima Y, Nozaki K, Nakanishi H, Yada E, Terai Y, Fueki K, Itaka K. Anti-Inflammatory Therapy for Temporomandibular Joint Osteoarthritis Using mRNA Medicine Encoding Interleukin-1 Receptor Antagonist. Pharmaceutics 2022; 14:1785. [PMID: 36145533 PMCID: PMC9505648 DOI: 10.3390/pharmaceutics14091785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Messenger RNA (mRNA) is an emerging drug modality for protein replacement therapy. As mRNA efficiently provides protein expression in post-mitotic cells without the risk of insertional mutagenesis, direct delivery of mRNA can be applied, not only as an alternative to gene therapy, but also for various common diseases such as osteoarthritis (OA). In this study, using an mRNA-encoding interleukin-1 receptor antagonist (IL-1Ra), we attempted anti-inflammatory therapy in a rat model of the temporomandibular joint (TMJ) OA, which causes long-lasting joint pain with chronic inflammation. For the intra-articular injection of mRNA, a polyplex nanomicelle, our original polymer-based carrier, was used to offer the advantage of excellent tissue penetration with few immunogenic responses. While the protein expression was transient, a single administration of IL-1Ra mRNA provided sustained pain relief and an inhibitory effect on OA progression for 4 weeks. The mRNA-loaded nanomicelles provided the encoded protein diffusely in the disc and articular cartilage without upregulation of the expression levels of the pro-inflammatory cytokines IL-6 and tumor necrosis factor-α (TNF-α). This proof-of-concept study demonstrates how anti-inflammatory proteins delivered by mRNA delivery using a polyplex nanomicelle could act to alleviate OA, stimulating the development of mRNA therapeutics.
Collapse
Affiliation(s)
- Jia Deng
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
- Department of Masticatory Function and Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan
| | - Yuta Fukushima
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Kosuke Nozaki
- Department of Advanced Prosthodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan
| | - Hideyuki Nakanishi
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Erica Yada
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
- NanoCarrier Co., Ltd., Tokyo 104-0031, Japan
| | - Yuki Terai
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Kenji Fueki
- Department of Masticatory Function and Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki 210-0821, Japan
| |
Collapse
|
16
|
Brain Dp140 alters glutamatergic transmission and social behaviour in the mdx52 mouse model of Duchenne muscular dystrophy. Prog Neurobiol 2022; 216:102288. [PMID: 35654209 DOI: 10.1016/j.pneurobio.2022.102288] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 04/06/2022] [Accepted: 05/25/2022] [Indexed: 12/23/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle disorder caused by DMD mutations and is characterized by neurobehavioural comorbidities due to dystrophin deficiency in the brain. The lack of Dp140, a dystrophin short isoform, is clinically associated with intellectual disability and autism spectrum disorders (ASDs), but its postnatal functional role is not well understood. To investigate synaptic function in the presence or absence of brain Dp140, we utilized two DMD mouse models, mdx23 and mdx52 mice, in which Dp140 is preserved or lacking, respectively. ASD-like behaviours were observed in pups and 8-week-old mdx52 mice lacking Dp140. Paired-pulse ratio of excitatory postsynaptic currents, glutamatergic vesicle number in basolateral amygdala neurons, and glutamatergic transmission in medial prefrontal cortex-basolateral amygdala projections were significantly reduced in mdx52 mice compared to those in wild-type and mdx23 mice. ASD-like behaviour and electrophysiological findings in mdx52 mice were ameliorated by restoration of Dp140 following intra-cerebroventricular injection of antisense oligonucleotide drug-induced exon 53 skipping or intra-basolateral amygdala administration of Dp140 mRNA-based drug. Our results implicate Dp140 in ASD-like behaviour via altered glutamatergic transmission in the basolateral amygdala of mdx52 mice.
Collapse
|
17
|
Qin S, Tang X, Chen Y, Chen K, Fan N, Xiao W, Zheng Q, Li G, Teng Y, Wu M, Song X. mRNA-based therapeutics: powerful and versatile tools to combat diseases. Signal Transduct Target Ther 2022; 7:166. [PMID: 35597779 PMCID: PMC9123296 DOI: 10.1038/s41392-022-01007-w] [Citation(s) in RCA: 219] [Impact Index Per Article: 109.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
The therapeutic use of messenger RNA (mRNA) has fueled great hope to combat a wide range of incurable diseases. Recent rapid advances in biotechnology and molecular medicine have enabled the production of almost any functional protein/peptide in the human body by introducing mRNA as a vaccine or therapeutic agent. This represents a rising precision medicine field with great promise for preventing and treating many intractable or genetic diseases. In addition, in vitro transcribed mRNA has achieved programmed production, which is more effective, faster in design and production, as well as more flexible and cost-effective than conventional approaches that may offer. Based on these extraordinary advantages, mRNA vaccines have the characteristics of the swiftest response to large-scale outbreaks of infectious diseases, such as the currently devastating pandemic COVID-19. It has always been the scientists’ desire to improve the stability, immunogenicity, translation efficiency, and delivery system to achieve efficient and safe delivery of mRNA. Excitingly, these scientific dreams have gradually been realized with the rapid, amazing achievements of molecular biology, RNA technology, vaccinology, and nanotechnology. In this review, we comprehensively describe mRNA-based therapeutics, including their principles, manufacture, application, effects, and shortcomings. We also highlight the importance of mRNA optimization and delivery systems in successful mRNA therapeutics and discuss the key challenges and opportunities in developing these tools into powerful and versatile tools to combat many genetic, infectious, cancer, and other refractory diseases.
Collapse
Affiliation(s)
- Shugang Qin
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoshan Tang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuting Chen
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kepan Chen
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Na Fan
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Xiao
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Zheng
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Guohong Li
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqing Teng
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Yoshinaga N, Uchida S, Dirisala A, Naito M, Koji K, Osada K, Cabral H, Kataoka K. Bridging mRNA and Polycation Using RNA Oligonucleotide Derivatives Improves the Robustness of Polyplex Micelles for Efficient mRNA Delivery. Adv Healthc Mater 2022; 11:e2102016. [PMID: 34913604 DOI: 10.1002/adhm.202102016] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/15/2021] [Indexed: 01/20/2023]
Abstract
Polyplex for messenger RNA (mRNA) delivery requires strong yet reversible association between mRNA and polycation for extracellular robustness and selective intracellular disintegration. Herein, RNA oligonucleotide (OligoRNA) derivatives that bridge mRNA and polycation are developed to stabilize polyplex micelles (PMs). A set of the OligoRNAs introduced with a polyol moiety in their 5' end is designed to hybridize to fixed positions along mRNA strand. After PM preparation from the hybridized mRNA and poly(ethylene glycol)-polycation block copolymer derived with phenylboronic acid (PBA) moieties in its cationic segment, PBA moieties form reversible phenylboronate ester linkages with a polyol moiety at 5' end of OligoRNAs and a diol moiety at their 3' end ribose, in the PM core. The OligoRNAs work as a node to bridge ionically complexed mRNA and polycation, thereby improving PM stability against polyion exchange reaction and ribonuclease attack in extracellular environment. After cellular uptake, intracellular high concentration of adenosine triphosphate triggers the cleavage of phenylboronate ester linkages, resulting in mRNA release from PM. Ultimately, the PM provides efficient mRNA introduction in cultured cells and mouse lungs after intratracheal administration, demonstrating the potential of the bridging strategy in polyplex-based mRNA delivery.
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Department of Bioengineering Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo Bunkyo‐ku Tokyo 113‐8656 Japan
- Biomacromolecule Research Team RIKEN Center for Sustainable Resource Science 2‐1 Hirosawa, Wakoshi Saitama 351‐0198 Japan
| | - Satoshi Uchida
- Department of Bioengineering Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo Bunkyo‐ku Tokyo 113‐8656 Japan
- Medical Chemistry Graduate School of Medical Science Kyoto Prefectural University of Medicine 1‐5 Shimogamohangi‐cho Sakyo‐ku Kyoto 606‐0823 Japan
- Innovation Center of NanoMedicine (iCONM) Kanagawa Institute of Industrial Promotion 3‐25‐14 Tonomachi, Kawasaki‐ku Kawasaki 210‐0821 Japan
| | - Anjaneyulu Dirisala
- Innovation Center of NanoMedicine (iCONM) Kanagawa Institute of Industrial Promotion 3‐25‐14 Tonomachi, Kawasaki‐ku Kawasaki 210‐0821 Japan
| | - Mitsuru Naito
- Center for Disease Biology and Integrative Medicine Graduate School of Medicine The University of Tokyo 7‐3‐1 Hongo Bunkyo‐ku Tokyo 113‐0033 Japan
| | - Kyoko Koji
- Department of Bioengineering Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo Bunkyo‐ku Tokyo 113‐8656 Japan
| | - Kensuke Osada
- Institute for Quantum Medical Science National Institutes for Quantum Science and Technology (QST) 4‐9‐1 Anagawa, Inage‐ku Chiba‐shi Chiba 263‐8555 Japan
| | - Horacio Cabral
- Department of Bioengineering Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo Bunkyo‐ku Tokyo 113‐8656 Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM) Kanagawa Institute of Industrial Promotion 3‐25‐14 Tonomachi, Kawasaki‐ku Kawasaki 210‐0821 Japan
| |
Collapse
|
19
|
Dirisala A, Uchida S, Li J, Van Guyse JFR, Hayashi K, Vummaleti SVC, Kaur S, Mochida Y, Fukushima S, Kataoka K. Effective mRNA Protection by Poly(l-ornithine) Synergizes with Endosomal Escape Functionality of a Charge-Conversion Polymer toward Maximizing mRNA Introduction Efficiency. Macromol Rapid Commun 2022; 43:e2100754. [PMID: 35286740 DOI: 10.1002/marc.202100754] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/12/2022] [Indexed: 12/13/2022]
Abstract
For efficient delivery of messenger (m)RNA, delivery carriers need two major functions: protecting mRNA from nucleases and translocating mRNA from endolysosomes to the cytoplasm. Herein, these two complementary functionalities are integrated into a single polyplex by fine-tuning the catiomer chemical structure and incorporating the endosomal escape modality. The effect of the methylene spacer length on the catiomer side chain is evaluated by comparing poly(l-lysine) (PLL) with a tetramethylene spacer and poly(L-ornithine) (PLO) with a trimethylene spacer. Noteworthily, the nuclease stability of the mRNA/catiomer polyplexes is largely affected by the difference in one methylene group, with PLO/mRNA polyplex showing enhanced stability compared to PLL/mRNA polyplex. To introduce the endosomal escape function, the PLO/mRNA polyplex is wrapped with a charge-conversion polymer (CCP), which is negatively charged at extracellular pH but turns positive at endosomal acidic pH to disrupt the endosomal membrane. Compared to the parent PLO/mRNA polyplex, CCP facilitated the endosomal escape of the polyplex in cultured cells to improve the protein expression efficiency from mRNA by approximately 80-fold. Collectively, this system synergizes the protective effect of PLO against nucleases and the endosomal escape capability of CCP in mRNA delivery.
Collapse
Affiliation(s)
- Anjaneyulu Dirisala
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.,Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto, 606-0823, Japan
| | - Junjie Li
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Joachim F R Van Guyse
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Kotaro Hayashi
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Sai V C Vummaleti
- Institute of High Performance Computing Agency for Science, Technology and Research, 1 Fusionopolis Way, 16-6 Connexis, 138632, Singapore
| | - Sarandeep Kaur
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Johannisallee 29, Leipzig, 04103, Germany
| | - Yuki Mochida
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Shigeto Fukushima
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kanagawa Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| |
Collapse
|
20
|
Pezzotti G, Zhu W, Terai Y, Marin E, Boschetto F, Kawamoto K, Itaka K. Raman spectroscopic insight into osteoarthritic cartilage regeneration by mRNA therapeutics encoding cartilage-anabolic transcription factor Runx1. Mater Today Bio 2022; 13:100210. [PMID: 35281370 PMCID: PMC8913780 DOI: 10.1016/j.mtbio.2022.100210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/18/2022] [Accepted: 01/28/2022] [Indexed: 11/05/2022] Open
Abstract
While joint arthroplasty remains nowadays the most popular option available to repair chronically degenerated osteoarthritic joints, possibilities are recently emerging for regeneration of damaged cartilage rather than its replacement with artificial biomaterials. This latter strategy could allow avoiding the quite intrusive surgical procedures associated with total joint replacement. Building upon this notion, we first apply Raman spectroscopy to characterize diseased cartilage in a mice model of instability-induced knee osteoarthritis (OA) upon medial collateral ligament (MCL) and medial meniscus (MM) transections. Then, we examine the same OA model after cartilage regeneration by means of messenger RNA (mRNA) delivery of a cartilage-anabolic runt-related transcription factor 1 (RUNX1). Raman spectroscopy is shown to substantiate at the molecular scale the therapeutic effect of the Runx1 mRNA cartilage regeneration approach. This study demonstrates how the Raman spectroscopic method could support and accelerate the development of new therapies for cartilage diseases.
Collapse
|
21
|
Steinle H, Weber J, Stoppelkamp S, Große-Berkenbusch K, Golombek S, Weber M, Canak-Ipek T, Trenz SM, Schlensak C, Avci-Adali M. Delivery of synthetic mRNAs for tissue regeneration. Adv Drug Deliv Rev 2021; 179:114007. [PMID: 34710530 DOI: 10.1016/j.addr.2021.114007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/03/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, nucleic acid-based therapeutics have gained increasing importance as novel treatment options for disease prevention and treatment. Synthetic messenger RNAs (mRNAs) are promising nucleic acid-based drugs to transiently express desired proteins that are missing or defective. Recently, synthetic mRNA-based vaccines encoding viral proteins have been approved for emergency use against COVID-19. Various types of vehicles, such as lipid nanoparticles (LNPs) and liposomes, are being investigated to enable the efficient uptake of mRNA molecules into desired cells. In addition, the introduction of novel chemical modifications into mRNAs increased the stability, enabled the modulation of nucleic acid-based drugs, and increased the efficiency of mRNA-based therapeutic approaches. In this review, novel and innovative strategies for the delivery of synthetic mRNA-based therapeutics for tissue regeneration are discussed. Moreover, with this review, we aim to highlight the versatility of synthetic mRNA molecules for various applications in the field of regenerative medicine and also discuss translational challenges and required improvements for mRNA-based drugs.
Collapse
Affiliation(s)
- Heidrun Steinle
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Josefin Weber
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sandra Stoppelkamp
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Katharina Große-Berkenbusch
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sonia Golombek
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Marbod Weber
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Tuba Canak-Ipek
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Sarah-Maria Trenz
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Christian Schlensak
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Meltem Avci-Adali
- University Hospital Tuebingen, Department of Thoracic and Cardiovascular Surgery, Calwerstraße 7/1, 72076 Tuebingen, Germany.
| |
Collapse
|
22
|
Nakanishi H. Protein-Based Systems for Translational Regulation of Synthetic mRNAs in Mammalian Cells. Life (Basel) 2021; 11:life11111192. [PMID: 34833067 PMCID: PMC8621430 DOI: 10.3390/life11111192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
Synthetic mRNAs, which are produced by in vitro transcription, have been recently attracting attention because they can express any transgenes without the risk of insertional mutagenesis. Although current synthetic mRNA medicine is not designed for spatiotemporal or cell-selective regulation, many preclinical studies have developed the systems for the translational regulation of synthetic mRNAs. Such translational regulation systems will cope with high efficacy and low adverse effects by producing the appropriate amount of therapeutic proteins, depending on the context. Protein-based regulation is one of the most promising approaches for the translational regulation of synthetic mRNAs. As synthetic mRNAs can encode not only output proteins but also regulator proteins, all components of protein-based regulation systems can be delivered as synthetic mRNAs. In addition, in the protein-based regulation systems, the output protein can be utilized as the input for the subsequent regulation to construct multi-layered gene circuits, which enable complex and sophisticated regulation. In this review, I introduce what types of proteins have been used for translational regulation, how to combine them, and how to design effective gene circuits.
Collapse
Affiliation(s)
- Hideyuki Nakanishi
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| |
Collapse
|
23
|
Delehedde C, Even L, Midoux P, Pichon C, Perche F. Intracellular Routing and Recognition of Lipid-Based mRNA Nanoparticles. Pharmaceutics 2021; 13:pharmaceutics13070945. [PMID: 34202584 PMCID: PMC8308975 DOI: 10.3390/pharmaceutics13070945] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/07/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Messenger RNA (mRNA) is being extensively used in gene therapy and vaccination due to its safety over DNA, in the following ways: its lack of integration risk, cytoplasmic expression, and transient expression compatible with fine regulations. However, clinical applications of mRNA are limited by its fast degradation by nucleases, and the activation of detrimental immune responses. Advances in mRNA applications, with the recent approval of COVID-19 vaccines, were fueled by optimization of the mRNA sequence and the development of mRNA delivery systems. Although delivery systems and mRNA sequence optimization have been abundantly reviewed, understanding of the intracellular processing of mRNA is mandatory to improve its applications. We will focus on lipid nanoparticles (LNPs) as they are the most advanced nanocarriers for the delivery of mRNA. Here, we will review how mRNA therapeutic potency can be affected by its interactions with cellular proteins and intracellular distribution.
Collapse
Affiliation(s)
- Christophe Delehedde
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
- Sanofi R&D, Integrated Drug Discovery, 91385 Chilly-Mazarin, France;
| | - Luc Even
- Sanofi R&D, Integrated Drug Discovery, 91385 Chilly-Mazarin, France;
| | - Patrick Midoux
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
| | - Chantal Pichon
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
- Correspondence: (C.P.); (F.P.); Tel.: +33-2-3825-5595 (C.P.); Tel.: +33-2-3825-5544 (F.P.)
| | - Federico Perche
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR4301, Rue Charles Sadron, 45071 Orléans, France; (C.D.); (P.M.)
- Correspondence: (C.P.); (F.P.); Tel.: +33-2-3825-5595 (C.P.); Tel.: +33-2-3825-5544 (F.P.)
| |
Collapse
|
24
|
Yoshinaga N, Naito M, Tachihara Y, Boonstra E, Osada K, Cabral H, Uchida S. PEGylation of mRNA by Hybridization of Complementary PEG-RNA Oligonucleotides Stabilizes mRNA without Using Cationic Materials. Pharmaceutics 2021; 13:800. [PMID: 34071840 PMCID: PMC8227728 DOI: 10.3390/pharmaceutics13060800] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Messenger RNA (mRNA) delivery strategies are required to protect biologically fragile mRNA from ribonuclease (RNase) attacks to achieve efficient therapeutic protein expression. To tackle this issue, most mRNA delivery systems have used cationic components, which form electrostatically driven complexes with mRNA and shield encapsulated mRNA strands. However, cationic materials interact with anionic biomacromolecules in physiological environments, which leads to unspecific reactions and toxicities. To circumvent this issue of cation-based approaches, herein, we propose a cation-free delivery strategy by hybridization of PEGylated RNA oligonucleotides with mRNA. The PEG strands on the mRNA sterically and electrostatically shielded the mRNA, improving mRNA nuclease stability 15-fold after serum incubation compared with unhybridized mRNA. Eventually, the PEGylated mRNA induced nearly 20-fold higher efficiency of reporter protein expression than unhybridized mRNA in cultured cells. This study provides a platform to establish a safe and efficient cation-free mRNA delivery system.
Collapse
Grants
- 20H04524 the Ministry of Education, Culture, Sports, Science and Technology, Japan (MEXT)
- 18K03529 the Ministry of Education, Culture, Sports, Science and Technology, Japan (MEXT)
- JP18K19901 the Ministry of Education, Culture, Sports, Science and Technology, Japan (MEXT)
- JP18ae0201009 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
- RIKEN Center for Sustainable Resource Science, Wako 351-0198, Saitama, Japan
| | - Mitsuru Naito
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan;
| | - Yoshihiro Tachihara
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
| | - Eger Boonstra
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
| | - Kensuke Osada
- National Institute of Radiological Science, 4-9-1 Anagawa, Inage-ku, Chiba-shi 236-8555, Chiba, Japan;
| | - Horacio Cabral
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
| | - Satoshi Uchida
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Inamori Memorial Building, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| |
Collapse
|
25
|
Uchida S. [Messenger RNA for Cell Editing -Disease Treatment and Cell Transplantation Using Pro-survival mRNA]. YAKUGAKU ZASSHI 2021; 141:655-659. [PMID: 33952747 DOI: 10.1248/yakushi.20-00219-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vigorous efforts are being made to manipulate cellular functions in a desirable manner for biomedical purposes. Recent advances in platform technologies have made cell editing achievable; this includes generation of induced pluripotent stem cells and chimeric antigen receptor T cells, as well as direct cell reprogramming. mRNA, as compared to DNA, is an excellent tool for potentiating cell editing technologies, owing to its distinct properties in gene introduction. Herein, hepatocytes were edited ex vivo and in vivo, by introducing pro-survival mRNA, to be resistant to cell death. DNA-based introduction of pro-survival gene poses safety concerns due to its genomic integration, as prolonged and uncontrolled expression of pro-survival proteins after the integration may promote cancer. In contrast, mRNA lacks such a risk. Moreover, mRNA-based introduction of Bcl-2, a pro-survival factor, was more effective in preventing the death of cultured hepatocytes than Bcl-2 plasmid DNA (pDNA) introduction. Mechanistically, mRNA induced protein expression in a larger percentage of the hepatocytes compared to pDNA, presumably because the process of pDNA nuclear entry in transfection is challenging. In hepatocyte transplantation to mouse liver, ex vivo introduction of Bcl-2 mRNA significantly improved the engraftment efficiency of the hepatocytes, leading to successful functional support of the liver in a mouse model of chronic hepatitis. Furthermore, in vivo administration of Bcl-2 mRNA exhibited an anti-apoptotic effect on the hepatocytes of a mouse model of fulminant hepatitis. These results demonstrate the potential advantages of mRNA introduction over DNA introduction in cell editing.
Collapse
Affiliation(s)
- Satoshi Uchida
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine.,Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion
| |
Collapse
|
26
|
Abbasi S, Uchida S. Multifunctional Immunoadjuvants for Use in Minimalist Nucleic Acid Vaccines. Pharmaceutics 2021; 13:644. [PMID: 34062771 PMCID: PMC8147386 DOI: 10.3390/pharmaceutics13050644] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/25/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Subunit vaccines based on antigen-encoding nucleic acids have shown great promise for antigen-specific immunization against cancer and infectious diseases. Vaccines require immunostimulatory adjuvants to activate the innate immune system and trigger specific adaptive immune responses. However, the incorporation of immunoadjuvants into nonviral nucleic acid delivery systems often results in fairly complex structures that are difficult to mass-produce and characterize. In recent years, minimalist approaches have emerged to reduce the number of components used in vaccines. In these approaches, delivery materials, such as lipids and polymers, and/or pDNA/mRNA are designed to simultaneously possess several functionalities of immunostimulatory adjuvants. Such multifunctional immunoadjuvants encode antigens, encapsulate nucleic acids, and control their pharmacokinetic or cellular fate. Herein, we review a diverse class of multifunctional immunoadjuvants in nucleic acid subunit vaccines and provide a detailed description of their mechanisms of adjuvanticity and induction of specific immune responses.
Collapse
Affiliation(s)
- Saed Abbasi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| |
Collapse
|
27
|
Patel P, Ibrahim NM, Cheng K. The Importance of Apparent pKa in the Development of Nanoparticles Encapsulating siRNA and mRNA. Trends Pharmacol Sci 2021; 42:448-460. [PMID: 33875229 DOI: 10.1016/j.tips.2021.03.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 02/08/2023]
Abstract
Polymer and lipid nanoparticles have been extensively used as carriers to address the biological barriers encountered in siRNA and mRNA delivery. We summarize the crucial role of nanoparticle charge and ionizability in complexing RNAs, binding to biological components, escaping from the endosome, and releasing RNAs into the cytoplasm. We highlight the significant impact of the apparent pKa of nanoparticles on their efficacy and toxicity, and the importance of optimizing pKa in the development of lead formulations for RNAs. We also discuss the feasibility of fine-tuning the pKa in nanoparticles and the applications of this approach in the optimization of delivery systems for RNAs.
Collapse
Affiliation(s)
- Pratikkumar Patel
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri - Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Nurudeen Mohammed Ibrahim
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri - Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri - Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA.
| |
Collapse
|
28
|
Abbasi S, Uchida S, Toh K, Tockary TA, Dirisala A, Hayashi K, Fukushima S, Kataoka K. Co-encapsulation of Cas9 mRNA and guide RNA in polyplex micelles enables genome editing in mouse brain. J Control Release 2021; 332:260-268. [PMID: 33647431 DOI: 10.1016/j.jconrel.2021.02.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/07/2021] [Accepted: 02/23/2021] [Indexed: 12/26/2022]
Abstract
Genome editing using CRISPR/Cas9 has attracted considerable attention for the treatment of genetic disorders and viral infections. Co-delivery of Cas9 mRNA and single guide (sg)RNA is a promising strategy to efficiently edit the genome of various cell types, including non-dividing cells, with minimal safety concerns. However, co-delivery of two RNA species with significantly different sizes, such as Cas9 mRNA (4.5 kb) and sgRNA (0.1 kb), is still challenging, especially in vivo. Here, we addressed this issue by using a PEGylated polyplex micelle (PM) condensing the RNA in its core. PM loading sgRNA alone released sgRNA at minimal dilution in buffer, while PM loading Cas9 mRNA alone was stable even at higher dilutions. Interestingly, co-encapsulating sgRNA with Cas9 mRNA in a single PM prevented sgRNA release upon dilution, which led to the enhanced tolerability of sgRNA against enzymatic degradation. Subsequently, PM with co-encapsulated RNA widely induced genome editing in parenchymal cells in the mouse brain, including neurons, astrocytes, and microglia, following intraparenchymal injection, at higher efficiency than that by co-delivery of PMs loaded with either Cas9 mRNA or sgRNA separately. To the best of our knowledge, this is the first report demonstrating the utility of RNA-based delivery of CRISPR/Cas9 in inducing genome editing in the brain parenchymal cells. Furthermore, the efficiency of genome editing using PMs was higher than using a non-PEGylated polyplex, due to the enhanced diffusion of PMs in the brain tissue. The results reported herein demonstrate the potential of using PMs to co-encapsulate Cas9 mRNA and sgRNA for in vivo genome editing.
Collapse
Affiliation(s)
- Saed Abbasi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan.
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Theofilus A Tockary
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Anjaneyulu Dirisala
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kotaro Hayashi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Shigeto Fukushima
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-1709, Japan.
| |
Collapse
|
29
|
Fukushima Y, Uchida S, Imai H, Nakatomi H, Kataoka K, Saito N, Itaka K. Treatment of ischemic neuronal death by introducing brain-derived neurotrophic factor mRNA using polyplex nanomicelle. Biomaterials 2021; 270:120681. [PMID: 33517206 DOI: 10.1016/j.biomaterials.2021.120681] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022]
Abstract
Ischemic neuronal death causes serious lifelong neurological deficits; however, there is no proven effective treatment that can prevent neuronal death after the ischemia. We investigated the feasibility of mRNA therapeutics for preventing the neuronal death in a rat model of transient global ischemia (TGI). By intraventricular administration of mRNA encoding brain-derived neurotrophic factor (BDNF) using a polymer-based carrier, polyplex nanomicelle, the mRNA significantly increased the survival rate of hippocampal neurons after TGI, with a rapid rise of BDNF in the hippocampus. Interestingly, mRNA administration on Day 2 after TGI provided significantly better survival rate than the administration immediately after TGI. Eventually, dosing twice on Day 2 and 5 exerted long-term therapeutic effects, which were confirmed by a Y-maze behavioral test demonstrating improved spatial memory compared with untreated rats on Day 20. Immunohistochemical analysis showed that astrocytes were chief targets of the BDNF mRNA-loaded nanomicelles, suggesting that the augmented BDNF secretion from astrocytes creates a supportive microenvironment for the neurons to tolerate changes caused by ischemic stresses, and terminate the process of progressive neuronal death after the ischemic attack. Overall, the unique mechanism of action of mRNA therapeutics provide a promising approach for preventing ischemic neuronal death.
Collapse
Affiliation(s)
- Yuta Fukushima
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 1010062, Tokyo, Japan; Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, 1138655, Tokyo, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 2100821, Kawasaki, Kanagawa, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 2100821, Kawasaki, Kanagawa, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 1138656, Tokyo, Japan
| | - Hideaki Imai
- Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, 1138655, Tokyo, Japan
| | - Hirofumi Nakatomi
- Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, 1138655, Tokyo, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 2100821, Kawasaki, Kanagawa, Japan; Institute for Future Initiatives, The University of Tokyo, 1130033, Tokyo, Japan
| | - Nobuhito Saito
- Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, 1138655, Tokyo, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 1010062, Tokyo, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 2100821, Kawasaki, Kanagawa, Japan.
| |
Collapse
|
30
|
van den Berg AIS, Yun CO, Schiffelers RM, Hennink WE. Polymeric delivery systems for nucleic acid therapeutics: Approaching the clinic. J Control Release 2021; 331:121-141. [PMID: 33453339 DOI: 10.1016/j.jconrel.2021.01.014] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
Gene therapy using nucleic acids has many clinical applications for the treatment of diseases with a genetic origin as well as for the development of innovative vaccine formulations. Since nucleic acids in their free form are rapidly degraded by nucleases present in extracellular matrices, have poor pharmacokinetics and hardly pass cellular membranes, carrier systems are required. Suitable carriers that protect the nucleic acid payload against enzymatic attack, prolong circulation time after systemic administration and assist in cellular binding and internalization are needed to develop nucleic acid based drug products. Viral vectors have been investigated and are also clinically used as delivery vehicles. However, some major drawbacks are associated with their use. Therefore there has been substantial attention on the use of non-viral carrier systems based on cationic lipids and polymers. This review focuses on the properties of polymer-based nucleic acid formulations, also referred as polyplexes. Different polymeric systems are summarized, and the cellular barriers polyplexes encounter and ways to tackle these are discussed. Finally attention is given to the clinical status of non-viral nucleic acid formulations.
Collapse
Affiliation(s)
- Annette I S van den Berg
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| | - Chae-Ok Yun
- Institute of Nano Science and Technology, Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584, CG, Utrecht, the Netherlands.
| |
Collapse
|
31
|
Yoshinaga N, Uchida S, Dirisala A, Naito M, Osada K, Cabral H, Kataoka K. mRNA loading into ATP-responsive polyplex micelles with optimal density of phenylboronate ester crosslinking to balance robustness in the biological milieu and intracellular translational efficiency. J Control Release 2020; 330:317-328. [PMID: 33359053 DOI: 10.1016/j.jconrel.2020.12.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023]
Abstract
Carriers for messenger RNA (mRNA) delivery require propensities to protect the mRNA from enzymatic degradation and to selectively release mRNA in the cytosol for smooth mRNA translation. To meet these requirements, we designed mRNA-loaded polyplex micelles (PMs) with ATP-responsive crosslinking in the inner core by complexing mRNA with poly(ethylene glycol)-polycation block copolymers derivatized with phenylboronic acid and polyol groups, which form crosslinking structures via spontaneous phenylboronate ester formation. PMs thus prepared are tolerable against enzymatic attack and, in turn, disintegrate in the cytosol to release mRNA when triggered by the cleavage of phenylboronate ester linkages in response to elevated ATP concentration. Two structural factors of the PM, including (i) the introduction ratios of phenylboronate ester crosslinkers and (ii) the structure and protonation degree of amino groups in the polycation segment, are critical for maximizing protein expression in cultured cells due to the optimized balance between the robustness in the biological milieu and the ATP-responsive mRNA release in the cytosol. The optimal PM formulation was further stabilized by installing cholesterol moieties into both the mRNA and ω-end of the block copolymer to elicit longevity in blood circulation after intravenous injection.
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.
| | - Anjaneyulu Dirisala
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Mitsuru Naito
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kensuke Osada
- Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
32
|
Zhang N, Lin J, Chin JS, Zhang K, Chew SY. A laser microdissection-based axotomy model incorporating the use of biomimicking fiber scaffolds reveals that microRNAs promote axon regeneration over long injury distances. Biomater Sci 2020; 8:6286-6300. [PMID: 33020773 DOI: 10.1039/d0bm01380c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The regeneration of injured neurons over long injury distances remains suboptimal. In order to successfully stimulate nerve regrowth, potent biomolecules are necessary. Furthermore, reproducible and translatable methods to test the potency of candidate drugs for enhancing nerve regeneration over long axotomy distances are also needed. To address these issues, we report a novel laser microdissection-based axotomy model that involves the use of biomimicking aligned fiber substrates to precisely control neuronal axotomy distances. Correspondingly, we demonstrate that in the absence of therapeutics, dorsal root ganglion (DRG) explants (consisting of neurons) axotomized within short distances from the main cell somas regenerated significantly longer than axons that were injured more distally (p < 0.05). However, when treated with a cocktail of microRNAs (miR-132/miR-222/miR-431), robust neurite outgrowth was observed (p < 0.05). Specifically, microRNA treatment promoted neurite outgrowth by ∼2.2-fold as compared to untreated cells and this enhancement was more significant under the less conducive regeneration condition of a long axotomy distance (i.e. 1000 μm from the cell soma). Besides that, we demonstrated that the treatment of miR-132/miR-222/miR-431 led to a longer length of nerve regeneration as well as a bigger nerve extension area after sciatic nerve transection injury. These results indicate that distance effects on axonal regrowth may be overcome by the effects of microRNAs and that these microRNAs may serve as promising therapeutics for nerve injury treatment.
Collapse
Affiliation(s)
- Na Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | | | | | | | | |
Collapse
|
33
|
Designing peptide nanoparticles for efficient brain delivery. Adv Drug Deliv Rev 2020; 160:52-77. [PMID: 33031897 DOI: 10.1016/j.addr.2020.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
The targeted delivery of therapeutic compounds to the brain is arguably the most significant open problem in drug delivery today. Nanoparticles (NPs) based on peptides and designed using the emerging principles of molecular engineering show enormous promise in overcoming many of the barriers to brain delivery faced by NPs made of more traditional materials. However, shortcomings in our understanding of peptide self-assembly and blood-brain barrier (BBB) transport mechanisms pose significant obstacles to progress in this area. In this review, we discuss recent work in engineering peptide nanocarriers for the delivery of therapeutic compounds to the brain: from synthesis, to self-assembly, to in vivo studies, as well as discussing in detail the biological hurdles that a nanoparticle must overcome to reach the brain.
Collapse
|
34
|
Abstract
Messenger RNA (mRNA) has immense potential for developing a wide range of therapies, including immunotherapy and protein replacement. As mRNA presents no risk of integration into the host genome and does not require nuclear entry for transfection, which allows protein production even in nondividing cells, mRNA-based approaches can be envisioned as safe and practical therapeutic strategies. Nevertheless, mRNA presents unfavorable characteristics, such as large size, immunogenicity, limited cellular uptake, and sensitivity to enzymatic degradation, which hinder its use as a therapeutic agent. While mRNA stability and immunogenicity have been ameliorated by direct modifications on the mRNA structure, further improvements in mRNA delivery are still needed for promoting its activity in biological settings. In this regard, nanomedicine has shown the ability for spatiotemporally controlling the function of a myriad of bioactive agents in vivo. Direct engineering of nanomedicine structures for loading, protecting, and releasing mRNA and navigating in biological environments can then be applied for promoting mRNA translation toward the development of effective treatments. Here, we review recent approaches aimed at enhancing mRNA function and its delivery through nanomedicines, with particular emphasis on their applications and eventual clinical translation.
Collapse
Affiliation(s)
- Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki 210-0821, Japan
| | - Federico Perche
- Centre de Biophysique Moléculaire, UPR4301 CNRS Rue Charles Sadron Orléans, Orléans 45071 Cedex 02, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, UPR4301 CNRS Rue Charles Sadron Orléans, Orléans 45071 Cedex 02, France.,Faculty of Sciences and Techniques, University of Orléans, Orléans 45071 Cedex 02, France
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki 210-0821, Japan
| |
Collapse
|
35
|
Lei S, Zhang X, Men K, Gao Y, Yang X, Wu S, Duan X, Wei Y, Tong R. Efficient Colorectal Cancer Gene Therapy with IL-15 mRNA Nanoformulation. Mol Pharm 2020; 17:3378-3391. [PMID: 32787272 DOI: 10.1021/acs.molpharmaceut.0c00451] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immunogene therapy is a novel method for the treatment of colorectal cancer. Cytokine IL-15 has exhibited therapeutic anticancer potential due to its immune-stimulation property. However, conventional IL-15-based cancer gene therapy studies have been performed using the plasmid DNA form, which has potential shortcomings including weak delivery efficiency and backbone effect. In this study, an IL-15 immunogene therapy study for colon cancer using in vitro transcript mRNA is described. A protamine/liposome system (CLPP) is developed to provide efficient condensation and delivery capacity for in vivo mRNA transportation. They demonstrated that the prepared CLPP system could deliver the IL-15-encoding mRNA into C26 cells with high efficacy. The secretory expressed IL-15 cytokine by the C26 cells successfully produced lymphocyte stimulation and triggered anticancer cytotoxicity upon cancer cells in vitro. Local or systemic administration of the CLPP/mIL-15 complex exhibited obvious inhibition effects on multiple C26 murine colon cancer models with inhibition rates of up to 70% in the C26 abdominal cavity metastasis tumor model, 55% in the subcutaneous model, and 69% in the pulmonary metastasis model, demonstrating high efficacy and safety. These results successfully demonstrated the high therapeutic potential of the CLPP/mIL-15 complex for colorectal cancer immunogene therapy.
Collapse
Affiliation(s)
- Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xueyan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Yan Gao
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xijing Yang
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Sisi Wu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, People's Republic of China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, People's Republic of China
| |
Collapse
|
36
|
Koji K, Yoshinaga N, Mochida Y, Hong T, Miyazaki T, Kataoka K, Osada K, Cabral H, Uchida S. Bundling of mRNA strands inside polyion complexes improves mRNA delivery efficiency in vitro and in vivo. Biomaterials 2020; 261:120332. [PMID: 32877764 DOI: 10.1016/j.biomaterials.2020.120332] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/14/2020] [Indexed: 01/08/2023]
Abstract
RNA nanotechnology has promise for developing mRNA carriers with enhanced physicochemical and functional properties. However, the potential synergy for mRNA delivery of RNA nanotechnology in cooperation with established carrier systems remains unknown. This study proposes a combinational system of RNA nanotechnology and mRNA polyplexes, by focusing on mRNA steric structure inside the polyplexes. Firstly, several mRNA strands are bundled through hybridization with RNA oligonucleotide crosslinkers to obtain tight mRNA structure, and then the bundled mRNA is mixed with poly(ethylene glycol) (PEG)-polycation block copolymers to prepare PEG-coated polyplex micelles (PMs). mRNA bundling results in highly condensed mRNA packaging inside PM core with dense PEG chains on the surface, thereby, improving PM stability against polyion exchange reaction and ribonuclease (RNase) attack. Importantly, such stabilization effects are attributed to bundled structure of mRNA rather than the increase in total mRNA amount encapsulated in the PMs, as encapsulation of long mRNA strands without bundling fails to improve PM stability. Consequently, PMs loading bundled mRNA exhibit enhanced stability in mouse blood circulation, and induce efficient protein expression in cultured cells and mouse brain.
Collapse
Affiliation(s)
- Kyoko Koji
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Naoto Yoshinaga
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Taehun Hong
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takuya Miyazaki
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Kanagawa Institute of Industrial Science and Technology, 705-1 Shimoimaizumi, Ebina, Kanagawa, 243-0435, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kensuke Osada
- Department of Molecular Imaging and Theranostics, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| | - Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| |
Collapse
|
37
|
Ke X, Shelton L, Hu Y, Zhu Y, Chow E, Tang H, Santos JL, Mao HQ. Surface-Functionalized PEGylated Nanoparticles Deliver Messenger RNA to Pulmonary Immune Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:35835-35844. [PMID: 32659078 PMCID: PMC9762545 DOI: 10.1021/acsami.0c08268] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanoparticles designed as messenger RNA (mRNA) carriers to deliver gene medicine have shown great potential to change the way lung disease states are managed. Controlling their delivery to the lung and the transgene expression in a specific population of cells remains a challenge. Here, we developed a series of nanoparticles with polyethylene glycol (PEG) corona prepared by condensing mRNA with PEG-grafted-polyethyleneimine (PEI-g-PEG) with different PEG terminal functional groups and grafting ratios. PEGylated nanoparticles (PEG grafting ratio was 0.5%) with amino or amino acid terminal groups showed the highest transgene expression levels in the lung following systemic administration, and cell profiling analysis indicated that pulmonary immune cells contributed to the majority of expression. We also showed that these nanoparticles can be prepared by the flash nanocomplexation method, which is a scalable and reproducible process, yielding lyophilizable nanoparticles that were stable for at least 4 months at -20 °C. These results suggest that these surface-functionalized PEGylated nanoparticles may serve as desirable carriers to deliver mRNA to the lung for pulmonary immunomodulation.
Collapse
Affiliation(s)
- Xiyu Ke
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lillie Shelton
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Emily Chow
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14850, USA
| | - Haoyu Tang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - José Luis Santos
- Biopharmaceuticals Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
38
|
Affiliation(s)
- Chaoyang Meng
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Xiangya Hospital of Central South University Changsha Hunan 410000 China
| | - Zhe Chen
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Xiangya Hospital of Central South University Changsha Hunan 410000 China
| | - Gang Li
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Thomas Welte
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Haifa Shen
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Cancer Center Houston Methodist Hospital Houston TX 77030 USA
- Department of Cell and Developmental Biology Weill Cornell Medical College New York NY 10065 USA
| |
Collapse
|
39
|
Fowler MJ, Cotter JD, Knight BE, Sevick-Muraca EM, Sandberg DI, Sirianni RW. Intrathecal drug delivery in the era of nanomedicine. Adv Drug Deliv Rev 2020; 165-166:77-95. [PMID: 32142739 DOI: 10.1016/j.addr.2020.02.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/17/2019] [Accepted: 02/28/2020] [Indexed: 12/23/2022]
Abstract
Administration of substances directly into the cerebrospinal fluid (CSF) that surrounds the brain and spinal cord is one approach that can circumvent the blood-brain barrier to enable drug delivery to the central nervous system (CNS). However, molecules that have been administered by intrathecal injection, which includes intraventricular, intracisternal, or lumbar locations, encounter new barriers within the subarachnoid space. These barriers include relatively high rates of turnover as CSF clears and potentially inadequate delivery to tissue or cellular targets. Nanomedicine could offer a solution. In contrast to the fate of freely administered drugs, nanomedicine systems can navigate the subarachnoid space to sustain delivery of therapeutic molecules, genes, and imaging agents within the CNS. Some evidence suggests that certain nanomedicine agents can reach the parenchyma following intrathecal administration. Here, we will address the preclinical and clinical use of intrathecal nanomedicine, including nanoparticles, microparticles, dendrimers, micelles, liposomes, polyplexes, and other colloidalal materials that function to alter the distribution of molecules in tissue. Our review forms a foundational understanding of drug delivery to the CSF that can be built upon to better engineer nanomedicine for intrathecal treatment of disease.
Collapse
Affiliation(s)
- M J Fowler
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School/University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - J D Cotter
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School/University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - B E Knight
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School/University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - E M Sevick-Muraca
- Brown Foundation Institute of Molecular Medicine, Center for Molecular Imaging, Houston, TX 77030, United States of America
| | - D I Sandberg
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School/University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America; Department of Pediatric Surgery, McGovern Medical School/University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America; Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, United States of America
| | - R W Sirianni
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School/University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America.
| |
Collapse
|
40
|
Effects of Tissue Pressure on Transgene Expression Characteristics via Renal Local Administration Routes from Ureter or Renal Artery in the Rat Kidney. Pharmaceutics 2020; 12:pharmaceutics12020114. [PMID: 32024046 PMCID: PMC7076412 DOI: 10.3390/pharmaceutics12020114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 11/17/2022] Open
Abstract
We previously developed a renal pressure-mediated transfection method (renal pressure method) as a kidney-specific in vivo gene delivery system. However, additional information on selecting other injection routes and applicable animals remains unclear. In this study, we selected renal arterial and ureteral injections as local administration routes and evaluated the characteristics of gene delivery such as efficacy, safety, and distribution in pressured kidney of rat. Immediately after the naked pDNA injection, via renal artery or ureter, the left kidney of the rat was pressured using a pressure controlling device. Transfection efficiency of the pressured kidney was about 100-fold higher than that of the injection only group in both administration routes. The optimal pressure intensity in the rat kidney was 1.2 N/cm2 for renal arterial injection and 0.9 N/cm2 for ureteral injection. We found that transgene expression site differs according to administration route: cortical fibroblasts and renal tubule in renal arterial injection and cortical and medullary tubule and medullary collecting duct in ureteral injection. This is the first report to demonstrate that the renal pressure method can also be effective, after renal arterial and ureteral injections, in rat kidney.
Collapse
|
41
|
Guevara ML, Persano S, Persano F. Lipid-Based Vectors for Therapeutic mRNA-Based Anti-Cancer Vaccines. Curr Pharm Des 2019; 25:1443-1454. [DOI: 10.2174/1381612825666190619150221] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023]
Abstract
Cancer vaccines have been widely explored as a key tool for effective cancer immunotherapy. Despite
a convincing rationale behind cancer vaccines, extensive past efforts were unsuccessful in mediating significantly
relevant anti-tumor activity in clinical studies. One of the major reasons for such poor outcome, among others, is
the low immunogenicity of more traditional vaccines, such as peptide-, protein- and DNA- based vaccines.
Recently, mRNA emerged as a promising alternative to traditional vaccine strategies due to its high immunogenicity,
suitability for large-scale and low-cost production, and superior safety profile. However, the clinical
application of mRNA-based anti-cancer vaccines has been limited by their instability and inefficient in vivo delivery.
Recent technological advances have now largely overcome these issues and lipid-based vectors have demonstrated
encouraging results as mRNA vaccine platforms against several types of cancers. This review intends to
provide a detailed overview of lipid-based vectors for the development of therapeutic mRNA-based anti-tumor
vaccines.
Collapse
Affiliation(s)
- Maria L. Guevara
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Stefano Persano
- Nanomaterials for Biomedical Applications, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Francesca Persano
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), University of Salento, Lecce, Italy
| |
Collapse
|
42
|
Biomaterial-based delivery systems of nucleic acid for regenerative research and regenerative therapy. Regen Ther 2019; 11:123-130. [PMID: 31338391 PMCID: PMC6626072 DOI: 10.1016/j.reth.2019.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/10/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Regenerative medicine is a new and promising medical method aiming at treating patients with defective or dysfunctional tissues by maintaining or enhancing the biological activity of cells. The development of biomaterial-based technologies, such as cell scaffolds and carriers for drug delivery system, are highly required to promote the regenerative research and regenerative therapy. Nucleic acids are one of the most feasible factors to efficiently modify the biological activity of cells. The effective and stable delivery of nucleic acids into cells is highly required to succeed in the modification. Biomaterials-based non-viral carriers or biological carriers, like exosomes, play an important role in the efficient delivery of nucleic acids. This review introduces the examples of regenerative research and regenerative therapy based on the delivery of nucleic acids with biomaterials technologies and emphasizes their importance to accomplish regenerative medicine. Modifying the activity of cells is important for regenerative medicine. Various nucleic acids regulate gene expression to modify the activity of cells. Intracellular delivery system is vital to the nucleic acids-based modification. Biomaterials are useful for the intracellular delivery of nucleic acids.
Collapse
Key Words
- Biomaterials
- CRISPR, clustered regularly interspaced short palindromic repeats
- Cas, CRISPR-associated systems
- Cell scaffold
- DDS, drug delivery system
- Drug delivery system
- ECM, extracellular matrix
- MSC, mesenchymal stem cells
- Nucleic acids
- PEG, polyethylene glycol
- PLGA, poly(d,l-lactic acid-co-glycolic acid)
- RISC, RNA-induced silencing complex
- RNAi, RNA interferince
- Regenerative research
- Regenerative therapy
- TALEN, transcription activator-like effector nuclease
- ZFN, zinc finger nucleases
- lncRNA, long non-coding RNA
- mRNA, messenger RNA
- miRNA, microRNA
- siRNA, small interfering RNA
Collapse
|
43
|
Enhancement of Motor Function Recovery after Spinal Cord Injury in Mice by Delivery of Brain-Derived Neurotrophic Factor mRNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:465-476. [PMID: 31344657 PMCID: PMC6658833 DOI: 10.1016/j.omtn.2019.06.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/27/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022]
Abstract
Spinal cord injury (SCI) is a debilitating condition that can cause impaired motor function or full paralysis. In the days to weeks following the initial mechanical injury to the spinal cord, inflammation and apoptosis can cause additional damage to the injured tissues. This secondary injury impairs recovery. Brain-derived neurotrophic factor is a secreted protein that has been shown to improve a variety of neurological conditions, including SCI, by promoting neuron survival and synaptic plasticity. This study treated a mouse model of contusion SCI using a single dose of brain-derived neurotrophic factor (BDNF) mRNA nanomicelles prepared with polyethylene glycol polyamino acid block copolymer directly injected into the injured tissue. BDNF levels in the injured spinal cord tissue were approximately doubled by mRNA treatment. Motor function was monitored using the Basso Mouse Scale and Noldus CatWalk Automated Gait Analysis System for 6 weeks post-injury. BDNF-treated mice showed improved motor function recovery, demonstrating the feasibility of mRNA delivery to treat SCI.
Collapse
|
44
|
Dirisala A, Uchida S, Tockary TA, Yoshinaga N, Li J, Osawa S, Gorantla L, Fukushima S, Osada K, Kataoka K. Precise tuning of disulphide crosslinking in mRNA polyplex micelles for optimising extracellular and intracellular nuclease tolerability. J Drug Target 2019; 27:670-680. [PMID: 30499743 DOI: 10.1080/1061186x.2018.1550646] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The major issues in messenger (m)RNA delivery are rapid mRNA degradation in the extracellular and intracellular spaces, which decreases the efficiency and duration for protein expression from mRNA. Stabilization of mRNA carriers using environment-responsive crosslinkings has promises to overcome these issues. Herein, we fine-tuned the structure of disulphide crosslinkings, which are selectively cleaved in the intracellular reductive environment, using the mRNA-loaded polyplex micelles (PMs) prepared from poly(ethylene glycol)-poly(L-lysine) (PEG-PLys) block copolymers, particularly by focussing on cationic charge density after the crosslinking. Primary amino groups in PLys segment were partially thiolated in two ways: One is to introduce 3-mercaptopropionyl (MP) groups via amide linkage, resulting in the decreased cationic charge density [PEG-PLys(MP)], and the other is the conversion of amino groups to 1-amidine-3-mercaptopropyl (AMP) groups with preserving cationic charge density [PEG-PLys(AMP)]. Compared to non-crosslinked and PEG-PLys(MP) PMs, PEG-PLys(AMP) PM attained tighter mRNA packaging in the PM core, thereby improving mRNA nuclease tolerability in serum and intracellular spaces, and providing enhanced protein expression in cultured cells at the optimal crosslinking density. These findings highlight the importance of cationic charge preservation in installing crosslinking moieties, providing a rationale for mRNA carrier design in the molecular level.
Collapse
Affiliation(s)
- Anjaneyulu Dirisala
- a Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion , Kawasaki , Kanagawa , Japan
| | - Satoshi Uchida
- a Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion , Kawasaki , Kanagawa , Japan.,b Department of Bioengineering , Graduate School of Engineering, The University of Tokyo , Bunkyo , Tokyo , Japan
| | - Theofilus A Tockary
- a Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion , Kawasaki , Kanagawa , Japan
| | - Naoto Yoshinaga
- b Department of Bioengineering , Graduate School of Engineering, The University of Tokyo , Bunkyo , Tokyo , Japan
| | - Junjie Li
- a Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion , Kawasaki , Kanagawa , Japan
| | - Shigehito Osawa
- a Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion , Kawasaki , Kanagawa , Japan
| | - Lahari Gorantla
- c Department of Bioengineering , College of Engineering, University of Washington , Washington , USA
| | - Shigeto Fukushima
- a Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion , Kawasaki , Kanagawa , Japan
| | - Kensuke Osada
- d National Institutes for Quantum and Radiology Science and Technology , Inage , Chiba , Japan
| | - Kazunori Kataoka
- a Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion , Kawasaki , Kanagawa , Japan.,e Policy Alternatives Research Institute, The University of Tokyo , Bunkyo , Tokyo , Japan
| |
Collapse
|
45
|
PEG-OligoRNA Hybridization of mRNA for Developing Sterically Stable Lipid Nanoparticles toward In Vivo Administration. Molecules 2019; 24:molecules24071303. [PMID: 30987102 PMCID: PMC6479949 DOI: 10.3390/molecules24071303] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/22/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023] Open
Abstract
Lipid nanoparticles (LNPs) exhibit high potential as carriers of messenger RNA (mRNA). However, the arduous preparation process of mRNA-loaded LNPs remains a huge obstacle for their widespread clinical application. Herein, we tackled this issue by mRNA PEGylation through hybridization with polyethylene glycol (PEG)-conjugated RNA oligonucleotides (PEG-OligoRNAs). Importantly, mRNA translational activity was preserved even after hybridization of 20 PEG-OligoRNAs per mRNA. The straightforward mixing of the PEGylated mRNA with lipofectamine LTX, a commercial lipid-based carrier, just by pipetting in aqueous solution, allowed the successful preparation of mRNA-loaded LNPs with a diameter below 100 nm, whereas the use of non-PEGylated mRNA provided large aggregates above 100- and 1000-nm. In vivo, LNPs prepared from PEG-OligoRNA-hybridized mRNA exhibited high structural stability in biological milieu, without forming detectable aggregates in mouse blood after intravenous injection. In contrast, LNPs from non-PEGylated mRNA formed several micrometer-sized aggregates in blood, leading to rapid clearance from blood circulation and deposition of the aggregates in lung capillaries. Our strategy of mRNA PEGylation was also versatile to prevent aggregation of another type of mRNA-loaded LNP, DOTAP/Chol liposomes. Together, our approach provides a simple and robust preparation method to LNPs for in vivo application.
Collapse
|
46
|
Lin CY, Crowley ST, Uchida S, Komaki Y, Kataoka K, Itaka K. Treatment of Intervertebral Disk Disease by the Administration of mRNA Encoding a Cartilage-Anabolic Transcription Factor. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:162-171. [PMID: 30889482 PMCID: PMC6424144 DOI: 10.1016/j.omtn.2019.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 02/10/2019] [Accepted: 02/16/2019] [Indexed: 12/27/2022]
Abstract
Intervertebral disk (IVD) degeneration is often associated with severity of lower back pain. IVD core is an avascular, highly hydrated tissue composed of type II collagen, glycosaminoglycans, and proteoglycans. The disk degeneration is not only a destruction of IVD structure but also is related to a disorder of the turnover of the disk matrix, leading the jelly-like IVD core to be replaced by fibrous components. Here we present a disease-modifying strategy for IVD degenerative diseases by direct regulation of the cells in the IVD using mRNA medicine, to alter the misbalanced homeostasis during disk degeneration. When mRNA encoding a cartilage-anabolic transcription factor, runt-related transcription factor-1, was administered to a rat model of coccygeal disk degeneration using a polyplex nanomicelle composed of polyethylene glycol-polyamino acid block copolymers and mRNA, the disk height was maintained to a significantly higher extent (≈81%) compared to saline control (69%), with prevention of fibrosis in the disk tissue. In addition, the use of nanomicelles effectively prevented inflammation, which was observed by injection of naked mRNA into the disk. This proof-of-concept study revealed that mRNA medicine has a potential for treating IVD degenerative diseases by introducing a cartilage-anabolic factor into the host cells, proposing a new therapeutic strategy using mRNA medicine.
Collapse
Affiliation(s)
- Chin-Yu Lin
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan; Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Samuel Thomas Crowley
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan; Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda-ku, Tokyo 101-0062, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo 113-8656, Japan
| | - Yuji Komaki
- Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Keiji Itaka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan; Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda-ku, Tokyo 101-0062, Japan.
| |
Collapse
|
47
|
Naito M, Otsu Y, Kamegawa R, Hayashi K, Uchida S, Kim HJ, Miyata K. Tunable nonenzymatic degradability of N-substituted polyaspartamide main chain by amine protonation and alkyl spacer length in side chains for enhanced messenger RNA transfection efficiency. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2019; 20:105-115. [PMID: 30787961 PMCID: PMC6374946 DOI: 10.1080/14686996.2019.1569818] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 06/09/2023]
Abstract
Degradability of polycations under physiological conditions is an attractive feature for their use in biomedical applications, such as the delivery of nucleic acids. This study aims to design polycations with tunable nonenzymatic degradability. A series of cationic N-substituted polyaspartamides were prepared to possess primary amine via various lengths of alkyl spacers in side chains. The degradation rate of each polyaspartamide derivative was determined by size exclusion chromatography under different pH conditions. The N-substituted polyaspartamide containing a 2-aminoethyl moiety in the side chain (PAsp(AE)) showed considerable degradability under physiological conditions (pH 7.4, 37 °C). In contrast, the N-substituted polyaspartamides bearing a longer alkyl spacer in the side chain, i.e. the 3-aminopropyl (PAsp(AP)) and 4-aminobutyl moieties (PAsp(AB)), more strongly suppressed degradation. Further, a positive correlation was observed between the degradation rate of N-substituted polyaspartamides and a deprotonation degree of primary amines in their side chains. Therefore, we conclude that the deprotonated primary amine in the side chain of N-substituted polyaspartamides can induce the degradation of the main chain through the activation of amide nitrogen in the side chain. When N-substituted polyaspartamides were utilized as a messenger RNA (mRNA) delivery vehicle via formation of polyion complexes (PICs), degradable PAsp(AE) elicited significantly higher mRNA expression efficiency in cultured cells compared to PAsp(AP) and PAsp(AB). The higher efficiency of PAsp(AE) might be due to the facilitated destabilization of PICs within the cells, directed toward mRNA release. Additionally, degradation of PAsp(AE) considerably reduced its cytotoxicity. Thus, our study highlights a useful design of well-defined cationic poly(amino acid)s with tunable nonenzymatic degradability.
Collapse
Affiliation(s)
- Mitsuru Naito
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuta Otsu
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Rimpei Kamegawa
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Kotaro Hayashi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Hyun Jin Kim
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
48
|
Uchida S, Kataoka K. Design concepts of polyplex micelles for in vivo therapeutic delivery of plasmid DNA and messenger RNA. J Biomed Mater Res A 2019; 107:978-990. [PMID: 30665262 DOI: 10.1002/jbm.a.36614] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
Abstract
Nonviral delivery of plasmid (p)DNA or messenger (m)RNA is a safe and promising therapeutic option to continuously supply therapeutic proteins into diseased tissues. In most cases of in vivo pDNA and mRNA delivery, these nucleic acids are loaded into carriers based on cationic polymers and/or lipids to prevent nuclease-mediated degradation before reaching target cells. The carriers should also evade host clearance mechanisms, including uptake by scavenger cells and filtration in the spleen. Installation of ligands onto the carriers can facilitate their rapid uptake into target cells. Meanwhile, carrier toxicity should be minimized not only for preventing undesirable adverse responses in patients, but also for preserving the function of transfected cells to exert therapeutic effects. Long-term progressive improvement of platform technologies has helped overcome most of these issues, though some still remain hindering the widespread clinical application of nonviral pDNA and mRNA delivery. This review discusses design concepts of nonviral carriers for in vivo delivery and the issues to be overcome, focusing especially on our own efforts using polyplex micelles. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 978-990, 2019.
Collapse
Affiliation(s)
- Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo 113-8656, Japan.,Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan.,Policy Alternatives Research Institute, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| |
Collapse
|
49
|
Yoshinaga N, Uchida S, Naito M, Osada K, Cabral H, Kataoka K. Induced packaging of mRNA into polyplex micelles by regulated hybridization with a small number of cholesteryl RNA oligonucleotides directed enhanced in vivo transfection. Biomaterials 2019; 197:255-267. [PMID: 30669016 DOI: 10.1016/j.biomaterials.2019.01.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/23/2018] [Accepted: 01/12/2019] [Indexed: 12/22/2022]
Abstract
There has been a progressive interest in the molecular design of polymers and lipids as synthetic carriers for targeting therapeutic mRNA in vivo with the ability to circumvent nuclease attack for treating intractable diseases. Herein, we developed a simple approach to attain one order of magnitude higher nuclease tolerability of mRNA through the formation of polyplex micelles (PMs) by combining ω-cholesteryl (ω-Chol)-poly (ethylene-glycol) (PEG)-polycation block copolymers with mRNA pre-hybridized with cholesterol (Chol)-tethered RNA oligonucleotides (Chol (+)-OligoRNA). Even one or a few short Chol (+)-OligoRNA anchors harboring along the 46-fold longer mRNA strand was sufficient to induce tight mRNA packaging in the PM core, as evidenced by Förster resonance energy transfer (FRET) measurement as well as by a longitudinal relaxation time (T1) measurement using NMR. These results suggest that Chol (+)-OligoRNA on mRNA strand serves as a node to attract ω-Chol moiety of the block copolymers to tighten the mRNA packaging in the PM core. These mRNA loaded PMs showed high tolerability against nuclease attack, and exerted appreciable protein translational activity in cultured cells without any inflammatory responses, achieved by shortening of the length of hybridizing Chol (+)-OligoRNAs to 17 nucleotides. Finally, the Chol (+)-OligoRNA-stabilized PM revealed efficient mRNA introduction into the mouse lungs via intratracheal administration, demonstrating in vivo utility of this formulation.
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| | - Mitsuru Naito
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kensuke Osada
- National Institute of Radiological Science, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
50
|
Chan LY, Khung YL, Lin CY. Preparation of Messenger RNA Nanomicelles via Non-Cytotoxic PEG-Polyamine Nanocomplex for Intracerebroventicular Delivery: A Proof-of-Concept Study in Mouse Models. NANOMATERIALS 2019; 9:nano9010067. [PMID: 30621291 PMCID: PMC6359661 DOI: 10.3390/nano9010067] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022]
Abstract
The specific delivery of messenger RNA (mRNA) is an excellent alternative to plasmid DNA, due to the latter’s potential risk for random integration into the host genome. In this study, we propose the use of specially tailored polyplex nanomicelles for the intravenous delivery of mRNA into the brain of mice. In brief, along the backbone of a polyaspartamide polymer that is terminated with a 42k Polyethylene glycol chain (PEG), aminoethylene-repeating groups (two, three, and four units, respectively) were conjugated to side-chains to promote electrostatic interactions with mRNA. This structural configuration would ultimately condense into a polyplex nanomicelle ranging between 24 and 34 nm, as was confirmed by transmission electron microscopy (TEM) and dynamic light scattering (DLS) while the chemistry of the synthesis was validated through NMR analysis. Subsequently, we hypothesized an important correlation pertaining to the role of hydrogen bonding between the interaction of polyamine and mRNA in due course. As a proof of concept, we encapsulated the luciferase (Luc2) mRNA as a reporter gene through in vitro transcription (IVT) and subsequently infused the polyplex nanomicelles into mouse brains via an intracerebroventricular (ICV) injection to bypass the blood–brain barriers (BBB). Data revealed that PEGylated polyplex nanomicelles possessing four repeating units of aminoethylene groups had exhibited the best Luc2 mRNA delivery efficiency with no significant immune response registered.
Collapse
Affiliation(s)
- Long Yi Chan
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | - Yit Lung Khung
- Department of Biological Science and Technology, China Medical University, No.91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | - Chin-Yu Lin
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung 40402, Taiwan.
- Department of Biological Science and Technology, China Medical University, No.91 Hsueh-Shih Road, Taichung 40402, Taiwan.
- Tsuzuki Institute for Traditional Medicine, Collage of Pharmacy, China Medical University, No.91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| |
Collapse
|