1
|
Hu J, Arvejeh PM, Bone S, Hett E, Marincola FM, Roh KH. Nanocarriers for cutting-edge cancer immunotherapies. J Transl Med 2025; 23:447. [PMID: 40234928 PMCID: PMC12001629 DOI: 10.1186/s12967-025-06435-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Cancer immunotherapy aims to harness the body's own immune system for effective and long-lasting elimination of malignant neoplastic tissues. Owing to the advance in understanding of cancer pathology and immunology, many novel strategies for enhancing immunological responses against various cancers have been successfully developed, and some have translated into excellent clinical outcomes. As one promising strategy for the next generation of immunotherapies, activating the multi-cellular network (MCN) within the tumor microenvironment (TME) to deploy multiple mechanisms of action (MOAs) has attracted significant attention. To achieve this effectively and safely, delivering multiple or pleiotropic therapeutic cargoes to the targeted sites of cancerous tissues, cells, and intracellular organelles is critical, for which numerous nanocarriers have been developed and leveraged. In this review, we first introduce therapeutic payloads categorized according to their predicted functions in cancer immunotherapy and their physicochemical structures and forms. Then, various nanocarriers, along with their unique characteristics, properties, advantages, and limitations, are introduced with notable recent applications in cancer immunotherapy. Following discussions on targeting strategies, a summary of each nanocarrier matching with suitable therapeutic cargoes is provided with comprehensive background information for designing cancer immunotherapy regimens.
Collapse
Affiliation(s)
- Joyce Hu
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | - Pooria M Arvejeh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sydney Bone
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Erik Hett
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | | | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
- Biotechnology Science and Engineering Program, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
| |
Collapse
|
2
|
He Z, Li F, Liu M, Liao J, Guo C. Porcine Reproductive and Respiratory Syndrome Virus: Challenges and Advances in Vaccine Development. Vaccines (Basel) 2025; 13:260. [PMID: 40266104 PMCID: PMC11945896 DOI: 10.3390/vaccines13030260] [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: 01/06/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 04/24/2025] Open
Abstract
Persistent infection of porcine reproductive and respiratory syndrome virus (PRRSV) significantly hampers both the quantity and quality of pork production in China. Although PRRSV is widely prevalent worldwide, the absence of effective vaccines has made it one of the major pathogens threatening the sustainable development of the global swine industry. Vaccination remains one of the most effective measures for controlling pathogen infections. However, the continuous genetic recombination and mutation of PRRSV demand more comprehensive strategies to address emerging threats, while ensuring the efficacy and safety of vaccines. This review provides an overview of the latest advances in PRRSV vaccine research, highlighting the importance of understanding the unique strengths and limitations of various vaccines in developing effective therapeutic approaches and vaccination strategies. Moreover, the development of adjuvants and antiviral drugs as adjuncts to combat PRRSV infection offers significant potential for enhancing disease control efforts. With the advancement of technologies such as proteolysis-targeting chimera (PROTAC) and mRNA, new avenues for controlling PRRSV and other pathogens are emerging, offering considerable hope. Ultimately, the goal of these vaccine developments is to alleviate the impact of PRRSV on animal health and the profitability of the swine industry.
Collapse
Affiliation(s)
| | | | | | | | - Chunhe Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (Z.H.); (F.L.); (M.L.); (J.L.)
| |
Collapse
|
3
|
Shah DD, Chorawala MR, Pandya AJ, Kothari N, Prajapati BG, Parekh PS. Advancing the Battle against Cystic Fibrosis: Stem Cell and Gene Therapy Insights. Curr Med Sci 2024; 44:1155-1174. [PMID: 39676146 DOI: 10.1007/s11596-024-2936-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/03/2024] [Indexed: 12/17/2024]
Abstract
Cystic fibrosis (CF) is a hereditary disorder characterized by mutations in the CFTR gene, leading to impaired chloride ion transport and subsequent thickening of mucus in various organs, particularly the lungs. Despite significant progress in CF management, current treatments focus mainly on symptom relief and do not address the underlying genetic defects. Stem cell and gene therapies present promising avenues for tackling CF at its root cause. Stem cells, including embryonic, induced pluripotent, mesenchymal, hematopoietic, and lung progenitor cells, offer regenerative potential by differentiating into specialized cells and modulating immune responses. Similarly, gene therapy aims to correct CFTR gene mutations by delivering functional copies of the gene into affected cells. Various approaches, such as viral and nonviral vectors, gene editing with CRISPR-Cas9, small interfering RNA (siRNA) therapy, and mRNA therapy, are being explored to achieve gene correction. Despite their potential, challenges such as safety concerns, ethical considerations, delivery system optimization, and long-term efficacy remain. This review provides a comprehensive overview of the current understanding of CF pathophysiology, the rationale for exploring stem cell and gene therapies, the types of therapies available, their mechanisms of action, and the challenges and future directions in the field. By addressing these challenges, stem cell and gene therapies hold promise for transforming CF management and improving the quality of life of affected individuals.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Aanshi J Pandya
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Nirjari Kothari
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana, 384012, India.
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
| | | |
Collapse
|
4
|
Kim S, Kim S, Kim S, Lee NE, Lee SH, Kim H, Lee H. Improvement of Therapeutic Effect via Inducing Non-Apoptotic Cell Death Using mRNA-Protection Nanocage. Adv Healthc Mater 2024; 13:e2400240. [PMID: 39081097 DOI: 10.1002/adhm.202400240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 07/22/2024] [Indexed: 10/30/2024]
Abstract
Necroptosis, a cell death mechanism with the characteristics of both apoptosis and necrosis, is proposed as a promising therapeutic approach for cancer therapy. Induction of necroptosis for cancer therapy may be possible through the regulation of the expression of a key factor gene receptor-interacting protein kinase-3 (RIPK3) via in vitro transcription (IVT) mRNA delivery. However, mRNA is susceptible to degradation and has a low delivery efficiency, which highlights the requirement of a proper delivery vehicle for intracellular delivery. Therefore, a new mRNA delivery system based on the nanostructured silica nanoparticles, termed mRNA-protective nanocage (mPN) has been developed. High-efficiency expression of RIPK3 and induction of necroptosis is achieved through delivery of RIPK3 IVT mRNA with mPN in vitro and in vivo models. Importantly, the mPN carrying RIPK3 mRNA distributed locally in tumors upon intravascular injection, and successfully induced necroptosis and immune cell infiltration, a hallmark of necroptosis. the suppression of tumor growth in a murine cancer model, demonstrating the synergistic effect of RIPK3 mRNA- and immune cell-mediated therapy is also observed. These findings suggest the potential for anticancer therapy through necroptosis induction and provide a strategy for the development of mRNA-based nanomedicine.
Collapse
Affiliation(s)
- Seoyoung Kim
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seongchan Kim
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, 52828, Republic of Korea
| | - Sojin Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Nan-Ee Lee
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Soo-Hwan Lee
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyunkyung Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Hyojin Lee
- Biomaterial Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- SKKU-KIST, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| |
Collapse
|
5
|
Krymchenko R, Coşar Kutluoğlu G, van Hout N, Manikowski D, Doberenz C, van Kuppevelt TH, Daamen WF. Elastogenesis in Focus: Navigating Elastic Fibers Synthesis for Advanced Dermal Biomaterial Formulation. Adv Healthc Mater 2024; 13:e2400484. [PMID: 38989717 DOI: 10.1002/adhm.202400484] [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: 02/07/2024] [Revised: 05/31/2024] [Indexed: 07/12/2024]
Abstract
Elastin, a fibrous extracellular matrix (ECM) protein, is the main component of elastic fibers that are involved in tissues' elasticity and resilience, enabling them to undergo reversible extensibility and to endure repetitive mechanical stress. After wounding, it is challenging to regenerate elastic fibers and biomaterials developed thus far have struggled to induce its biosynthesis. This review provides a comprehensive summary of elastic fibers synthesis at the cellular level and its implications for biomaterial formulation, with a particular focus on dermal substitutes. The review delves into the intricate process of elastogenesis by cells and investigates potential triggers for elastogenesis encompassing elastin-related compounds, ECM components, and other molecules for their potential role in inducing elastin formation. Understanding of the elastogenic processes is essential for developing biomaterials that trigger not only the synthesis of the elastin protein, but also the formation of a functional and branched elastic fiber network.
Collapse
Affiliation(s)
- Roman Krymchenko
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Gizem Coşar Kutluoğlu
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
- MedSkin Solutions Dr. Suwelack AG, 48727, Billerbeck, Germany
| | - Noor van Hout
- Department of Dermatology, Radboud university medical center, Nijmegen, 6525 GA, The Netherlands
| | | | | | - Toin H van Kuppevelt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Willeke F Daamen
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| |
Collapse
|
6
|
Cantero MJ, Bueloni B, Gonzalez Llamazares L, Fiore E, Lameroli L, Atorrasagasti C, Mazzolini G, Malvicini M, Bayo J, García MG. Modified mesenchymal stromal cells by in vitro transcribed mRNA: a therapeutic strategy for hepatocellular carcinoma. Stem Cell Res Ther 2024; 15:208. [PMID: 38992782 PMCID: PMC11241816 DOI: 10.1186/s13287-024-03806-0] [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: 12/29/2023] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) tropism for tumours allows their use as carriers of antitumoural factors and in vitro transcribed mRNA (IVT mRNA) is a promising tool for effective transient expression without insertional mutagenesis risk. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine with antitumor properties by stimulating the specific immune response. The aim of this work was to generate modified MSCs by IVT mRNA transfection to overexpress GM-CSF and determine their therapeutic effect alone or in combination with doxorubicin (Dox) in a murine model of hepatocellular carcinoma (HCC). METHODS DsRed or GM-CSF IVT mRNAs were generated from a cDNA template designed with specific primers followed by reverse transcription. Lipofectamine was used to transfect MSCs with DsRed (MSC/DsRed) or GM-CSF IVT mRNA (MSC/GM-CSF). Gene expression and cell surface markers were determined by flow cytometry. GM-CSF secretion was determined by ELISA. For in vitro experiments, the J774 macrophage line and bone marrow monocytes from mice were used to test GM-CSF function. An HCC model was developed by subcutaneous inoculation (s.c.) of Hepa129 cells into C3H/HeN mice. After s.c. injection of MSC/GM-CSF, Dox, or their combination, tumour size and mouse survival were evaluated. Tumour samples were collected for mRNA analysis and flow cytometry. RESULTS DsRed expression by MSCs was observed from 2 h to 15 days after IVT mRNA transfection. Tumour growth remained unaltered after the administration of DsRed-expressing MSCs in a murine model of HCC and MSCs expressing GM-CSF maintained their phenotypic characteristic and migration capability. GM-CSF secreted by modified MSCs induced the differentiation of murine monocytes to dendritic cells and promoted a proinflammatory phenotype in the J774 macrophage cell line. In vivo, MSC/GM-CSF in combination with Dox strongly reduced HCC tumour growth in C3H/HeN mice and extended mouse survival in comparison with individual treatments. In addition, the tumours in the MSC/GM-CSF + Dox treated group exhibited elevated expression of proinflammatory genes and increased infiltration of CD8 + T cells and macrophages. CONCLUSIONS Our results showed that IVT mRNA transfection is a suitable strategy for obtaining modified MSCs for therapeutic purposes. MSC/GM-CSF in combination with low doses of Dox led to a synergistic effect by increasing the proinflammatory tumour microenvironment, enhancing the antitumoural response in HCC.
Collapse
Affiliation(s)
- María José Cantero
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Barbara Bueloni
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lucrecia Gonzalez Llamazares
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Esteban Fiore
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lucia Lameroli
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Guillermo Mazzolini
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariana Malvicini
- Cancer Immunobiology Laboratory, IIMT, Universidad Austral - CONICET, Buenos Aires, Argentina
| | - Juan Bayo
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariana G García
- Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
7
|
Moradian H, Schwestka M, Roch T, Gossen M. Deconvolution of synthetic mRNA expression: Nucleoside chemistry alters translatability. Bioeng Transl Med 2024; 9:e10622. [PMID: 39036083 PMCID: PMC11256140 DOI: 10.1002/btm2.10622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/21/2023] [Accepted: 11/09/2023] [Indexed: 07/23/2024] Open
Abstract
Recent technological advances in the production of in vitro transcribed messenger RNA (IVT-mRNA) facilitate its clinical use as well as its application in basic research. In this regard, numerous chemical modifications, which are not naturally observed in endogenous mRNA, have been implemented primarily to address the issue of immunogenicity and improve its biological performance. However, recent findings suggested pronounced differences between expression levels of IVT-mRNAs with different nucleoside modifications in transfected cells. Given the multistep process of IVT-mRNA delivery and subsequent intracellular expression, it is unclear which step is influenced by IVT-mRNA chemistry. Here, we deconvolute this process and show that the nucleoside modification does not interfere with complexation of carriers, their physicochemical properties, and extracellular stability, as exemplified by selected modifications. The immediate effect of mRNA chemistry on the efficiency of ribosomal protein synthesis as a contributor to differences in expression was quantified by in vitro cell-free translation. Our results demonstrate that for the nucleoside modifications tested, translatability was the decisive step in determining overall protein production. Also of special importance for future work on rational selection of tailored synthetic mRNA chemistries, our findings set a workflow to identify potentially limiting, modification-dependent steps in the complex delivery process.
Collapse
Affiliation(s)
- Hanieh Moradian
- Institute of Active Polymers, Helmholtz‐Zentrum HereonTeltowGermany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
| | - Marko Schwestka
- Institute of Active Polymers, Helmholtz‐Zentrum HereonTeltowGermany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
| | - Toralf Roch
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
- CheckImmune GmbH, Campus Virchow KlinikumBerlinGermany
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz‐Zentrum HereonTeltowGermany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
| |
Collapse
|
8
|
Movahed F, Darzi S, Mahdavi P, Salih Mahdi M, Qutaiba B Allela O, Naji Sameer H, Adil M, Zarkhah H, Yasamineh S, Gholizadeh O. The potential use of therapeutics and prophylactic mRNA vaccines in human papillomavirus (HPV). Virol J 2024; 21:124. [PMID: 38822328 PMCID: PMC11143593 DOI: 10.1186/s12985-024-02397-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024] Open
Abstract
Cervical cancer (CC) and other malignant malignancies are acknowledged to be primarily caused by persistent human papillomavirus (HPV) infection. Historically, vaccinations against viruses that produce neutralizing antibodies unique to the virus have been an affordable way to manage viral diseases. CC risk is decreased, but not eliminated, by HPV vaccinations. Since vaccinations have been made available globally, almost 90% of HPV infections have been successfully avoided. On the lesions and diseases that are already present, however, no discernible treatment benefit has been shown. As a result, therapeutic vaccines that elicit immune responses mediated by cells are necessary for the treatment of established infections and cancers. mRNA vaccines possess remarkable potential in combating viral diseases and malignancy as a result of their superior industrial production, safety, and efficacy. Furthermore, considering the expeditiousness of production, the mRNA vaccine exhibits promise as a therapeutic approach targeting HPV. Given that the HPV-encoded early proteins, including oncoproteins E6 and E7, are consistently present in HPV-related cancers and pre-cancerous lesions and have crucial functions in the progression and persistence of HPV-related diseases, they serve as ideal targets for therapeutic HPV vaccines. The action mechanism of HPV and HPV-related cancer mRNA vaccines, their recent advancements in clinical trials, and the potential for their therapeutic applications are highlighted in this study, which also offers a quick summary of the present state of mRNA vaccines. Lastly, we highlight a few difficulties with mRNA HPV vaccination clinical practice and provide our thoughts on further advancements in this quickly changing sector. It is expected that mRNA vaccines will soon be produced quickly for clinical HPV prevention and treatment.
Collapse
Affiliation(s)
- Fatemeh Movahed
- Department of Gynecology and Obstetrics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Satinik Darzi
- Department Of Obstetrics and Gynecology, Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Parya Mahdavi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| | - Hasna Zarkhah
- Department of Obstetrics and Gynaecology, Tabriz University of Medical Siences, Tabriz, Iran.
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | | |
Collapse
|
9
|
Weber J, Linti C, Lörch C, Weber M, Andt M, Schlensak C, Wendel HP, Doser M, Avci-Adali M. Combination of melt-electrospun poly-ε-caprolactone scaffolds and hepatocyte-like cells from footprint-free hiPSCs to create 3D biohybrid constructs for liver tissue engineering. Sci Rep 2023; 13:22174. [PMID: 38092880 PMCID: PMC10719291 DOI: 10.1038/s41598-023-49117-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 12/04/2023] [Indexed: 12/17/2023] Open
Abstract
The liver is a vital organ with numerous functions, including metabolic functions, detoxification, and the synthesis of secretory proteins. The increasing prevalence of liver diseases requires the development of effective treatments, models, and regenerative approaches. The field of liver tissue engineering represents a significant advance in overcoming these challenges. In this study, 3D biohybrid constructs were created by combining hepatocyte-like cells (HLCs) derived from patient-specific footprint-free human induced pluripotent stem cells (hiPSCs) and 3D melt-electrospun poly-ε-caprolactone (PCL) scaffolds. First, a differentiation procedure was established to obtain autologous HCLs from hiPSCs reprogrammed from renal epithelial cells using self-replicating mRNA. The obtained cells expressed hepatocyte-specific markers and exhibited important hepatocyte functions, such as albumin synthesis, cytochrome P450 activity, glycogen storage, and indocyanine green metabolism. Biocompatible PCL scaffolds were fabricated by melt-electrospinning and seeded with pre-differentiated hepatoblasts, which uniformly attached to the fibers of the scaffolds and successfully matured into HLCs. The use of patient-specific, footprint-free hiPSC-derived HLCs represents a promising cell source for personalized liver regeneration strategies. In combination with biocompatible 3D scaffolds, this innovative approach has a broader range of applications spanning liver tissue engineering, drug testing and discovery, and disease modeling.
Collapse
Affiliation(s)
- Josefin Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076, Tuebingen, Germany
| | - Carsten Linti
- Biomedical Engineering, German Institutes of Textile and Fiber Research Denkendorf DITF, Körschtalstraße 26, 73770, Denkendorf, Germany
| | - Christiane Lörch
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076, Tuebingen, Germany
| | - Marbod Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076, Tuebingen, Germany
| | - Madelene Andt
- Biomedical Engineering, German Institutes of Textile and Fiber Research Denkendorf DITF, Körschtalstraße 26, 73770, Denkendorf, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076, Tuebingen, Germany
| | - Hans Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076, Tuebingen, Germany
| | - Michael Doser
- Biomedical Engineering, German Institutes of Textile and Fiber Research Denkendorf DITF, Körschtalstraße 26, 73770, Denkendorf, Germany
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076, Tuebingen, Germany.
| |
Collapse
|
10
|
Zhao Y, Gan L, Ke D, Chen Q, Fu Y. Mechanisms and research advances in mRNA antibody drug-mediated passive immunotherapy. J Transl Med 2023; 21:693. [PMID: 37794448 PMCID: PMC10552228 DOI: 10.1186/s12967-023-04553-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Antibody technology is widely used in the fields of biomedical and clinical therapies. Nonetheless, the complex in vitro expression of recombinant proteins, long production cycles, and harsh storage conditions have limited their applications in medicine, especially in clinical therapies. Recently, this dilemma has been overcome to a certain extent by the development of mRNA delivery systems, in which antibody-encoding mRNAs are enclosed in nanomaterials and delivered to the body. On entering the cytoplasm, the mRNAs immediately bind to ribosomes and undergo translation and post-translational modifications. This process produces monoclonal or bispecific antibodies that act directly on the patient. Additionally, it eliminates the cumbersome process of in vitro protein expression and extends the half-life of short-lived proteins, which significantly reduces the cost and duration of antibody production. This review focuses on the benefits and drawbacks of mRNA antibodies compared with the traditional in vitro expressed antibodies. In addition, it elucidates the progress of mRNA antibodies in the prevention of infectious diseases and oncology therapy.
Collapse
Affiliation(s)
- Yuxiang Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China
| | - Linchuan Gan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China
| | - Dangjin Ke
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China.
| | - Yajuan Fu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China.
| |
Collapse
|
11
|
Burdette D, Ci L, Shilliday B, Slauter R, Auerbach A, Kenney M, Almarsson Ö, Cheung E, Hendrick T. Systemic Exposure, Metabolism, and Elimination of [ 14C]-Labeled Amino Lipid, Lipid 5, after a Single Administration of mRNA Encapsulating Lipid Nanoparticles to Sprague-Dawley Rats. Drug Metab Dispos 2023; 51:804-812. [PMID: 37208185 DOI: 10.1124/dmd.122.001194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/17/2023] [Accepted: 04/05/2023] [Indexed: 05/21/2023] Open
Abstract
The emerging therapeutic modality of lipid nanoparticle (LNP)-encapsulated mRNAs has demonstrated promising clinical results when used as vaccines and is currently being tested in formulations for a wide range of targeted chronic disease treatments. These therapeutics are multicomponent assemblages of well-characterized naturally occurring molecules in addition to xenobiotic molecules, whose in vivo distributions are poorly understood. Here, the metabolic outcome and in vivo elimination of heptadecan-9-yl 8-((2-hydroxyethyl) (8-(nonyloxy)-8-oxooctyl)amino)octanoate (Lipid 5), a key xenobiotic amino lipid in LNP formulations, were assessed after intravenous administration of 14C-labeled Lipid 5 to Sprague-Dawley rats. Intact Lipid 5 was predominantly cleared from plasma within 10 hour after dosing, with only small quantities (<1% of 14C dose) of a single diacid metabolite detected after 10 hour. Lipid 5 was rapidly metabolized via ester hydrolysis into aliphatic alcohols and diacidic amino head group moieties, which were further metabolized via β-oxidation. Overall, >90% of the administered Lipid 5-derived 14C was recovered in urine (65%) and feces (35%), predominantly as oxidative metabolites, within 72 hour after dosing, indicating rapid renal and hepatic elimination. In vitro metabolite identification after incubation with human, nonhuman primate, and rat hepatocytes showed similar metabolites to those found in vivo. No meaningful differences were observed in Lipid 5 metabolism or elimination by sex. In conclusion, Lipid 5, a critical amino lipid component of LNPs for mRNA therapeutic delivery, showed minimal exposure, rapid metabolism, and near-complete elimination of 14C metabolites in rats. SIGNIFICANCE STATEMENT: Heptadecan-9-yl 8-((2-hydroxyethyl) (8-(nonyloxy)-8-oxooctyl)amino)octanoate (Lipid 5) is a key component of lipid nanoparticles used for the delivery of mRNA-based medicines; understanding the rates and routes of its clearance is crucial to assessing its long-term safety in lipid nanoparticle technology. This study conclusively established the rapid metabolism, and near-complete elimination of intravenously administered [14C]Lipid 5 in rats via both liver and kidney as oxidative metabolites derived from ester hydrolysis and subsequent β-oxidation.
Collapse
Affiliation(s)
- Douglas Burdette
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Lei Ci
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Barclay Shilliday
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Richard Slauter
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Andrew Auerbach
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Matthew Kenney
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Örn Almarsson
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Eugene Cheung
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| | - Tracy Hendrick
- Moderna, Inc., Cambridge, Massachusetts (D.B., L.C., A.A., M.K., E.C.); Charles River Laboratories, Mattawan, Michigan (B.S., R.S.); Duke University, Durham, North Carolina (T.H.); and Lyndra Therapeutics, Cambridge, Massachusetts (Ö.A.)
| |
Collapse
|
12
|
The potential of RNA-based therapy for kidney diseases. Pediatr Nephrol 2023; 38:327-344. [PMID: 35507149 PMCID: PMC9066145 DOI: 10.1007/s00467-021-05352-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 01/10/2023]
Abstract
Inherited kidney diseases (IKDs) are a large group of disorders affecting different nephron segments, many of which progress towards kidney failure due to the absence of curative therapies. With the current advances in genetic testing, the understanding of the molecular basis and pathophysiology of these disorders is increasing and reveals new potential therapeutic targets. RNA has revolutionized the world of molecular therapy and RNA-based therapeutics have started to emerge in the kidney field. To apply these therapies for inherited kidney disorders, several aspects require attention. First, the mRNA must be combined with a delivery vehicle that protects the oligonucleotides from degradation in the blood stream. Several types of delivery vehicles have been investigated, including lipid-based, peptide-based, and polymer-based ones. Currently, lipid nanoparticles are the most frequently used formulation for systemic siRNA and mRNA delivery. Second, while the glomerulus and tubules can be reached by charge- and/or size-selectivity, delivery vehicles can also be equipped with antibodies, antibody fragments, targeting peptides, carbohydrates or small molecules to actively target receptors on the proximal tubule epithelial cells, podocytes, mesangial cells or the glomerular endothelium. Furthermore, local injection strategies can circumvent the sequestration of RNA formulations in the liver and physical triggers can also enhance kidney-specific uptake. In this review, we provide an overview of current and potential future RNA-based therapies and targeting strategies that are in development for kidney diseases, with particular interest in inherited kidney disorders.
Collapse
|
13
|
Bartůňková J, Bloomfield M, Havlišová M, Klocperk A, Kubešová H, Podrazil M, Střížová Z, Šedivá A. News in immunology. VNITRNI LEKARSTVI 2023; 69:133-137. [PMID: 37072272 DOI: 10.36290/vnl.2023.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
The field of immunology has undergone a very significant development in recent decades, which has been reflected especially in the beginning of this millennium in significant advances in the understanding of the immune system and in the application of this knowledge in practice. The progress and acceleration of research and advances in the field of immunology was further prompt by the unexpected onset of the COVID-19 pandemic in 2020. The intense scientific work has not only led to the development of our understanding of the immune response to viruses, but also to the rapid conversion of this knowledge into practical pandemic management on a global scale, as exemplified by the development of vaccines against SARS-Cov-2 virus. The pandemic era has further contributed to the acceleration of the application of not only biological discoveries but also technological approaches into practical applications, such as use of advanced mathematics, computer science and, more recently, artificial intelligence which are all are adding to the advances that are significantly moving the field of immunology forward. In this communication, we present specific advances in particular areas of immunopathology, which are mainly allergy, immunodeficiency, immunity and infection, vaccination, autoimmune diseases and cancer immunology.
Collapse
|
14
|
Tschorn N, van Heuvel Y, Stitz J. Transgene Expression and Transposition Efficiency of Two-Component Sleeping Beauty Transposon Vector Systems Utilizing Plasmid or mRNA Encoding the Transposase. Mol Biotechnol 2022:10.1007/s12033-022-00642-6. [DOI: 10.1007/s12033-022-00642-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
AbstractThe use of two-component transposon plasmid vector systems, namely, a transposase construct and a donor vector carrying the gene of interest (GOI) can accelerate the development of recombinant cell lines. However, the undesired stable transfection of the transposase construct and the sustained expression of the enzyme can cause genetic instability due to the re-mobilization of the previously transposed donor vectors. Using a Sleeping Beauty-derived vector system, we established three recombinant cell pools and demonstrate stable integration of the transposase construct and sustained expression of the transposase over a period of 48 days. To provide an alternative approach, transcripts of the transposase gene were generated in vitro and co-transfected with donor vector plasmid at different ratios and mediating high GOI copy number integrations and expression levels. We anticipate that the use of transposase mRNA will foster further improvements in future cell line development processes.
Collapse
|
15
|
Li CY, Liang Z, Hu Y, Zhang H, Setiasabda KD, Li J, Ma S, Xia X, Kuang Y. Cytidine-containing tails robustly enhance and prolong protein production of synthetic mRNA in cell and in vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:300-310. [PMID: 36320322 PMCID: PMC9614650 DOI: 10.1016/j.omtn.2022.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Synthetic mRNAs are rising rapidly as alternative therapeutic agents for delivery of proteins. However, the practical use of synthetic mRNAs has been restricted by their low cellular stability as well as poor protein production efficiency. The key roles of poly(A) tail on mRNA biology inspire us to explore the optimization of tail sequence to overcome the aforementioned limitations. Here, the systematic substitution of non-A nucleotides in the tails revealed that cytidine-containing tails can substantially enhance the protein production rate and duration of synthetic mRNAs both in vitro and in vivo. Such C-containing tails shield synthetic mRNAs from deadenylase CCR4-NOT transcription complex, as the catalytic CNOT proteins, especially CNOT6L and CNOT7, have lower efficiency in trimming of cytidine. Consistently, these enhancement effects of C-containing tails were observed on all synthetic mRNAs tested and were independent of transfection reagents and cell types. As the C-containing tails can be used along with other mRNA enhancement technologies to synergically boost protein production, we believe that these tails can be broadly used on synthetic mRNAs to directly promote their clinical applications.
Collapse
Affiliation(s)
- Cheuk Yin Li
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Zhenghua Liang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Yaxin Hu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Hongxia Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Kharis Daniel Setiasabda
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jiawei Li
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518057, China
| | - Shaohua Ma
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong 518057, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China,HKUST Shenzhen Research Institute, Shenzhen, Guangdong 518057, China,Corresponding author Yi Kuang, Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, Hong Kong.
| |
Collapse
|
16
|
Wang WC, Sayedahmed EE, Sambhara S, Mittal SK. Progress towards the Development of a Universal Influenza Vaccine. Viruses 2022; 14:v14081684. [PMID: 36016306 PMCID: PMC9415875 DOI: 10.3390/v14081684] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 11/21/2022] Open
Abstract
Influenza viruses are responsible for millions of cases globally and significantly threaten public health. Since pandemic and zoonotic influenza viruses have emerged in the last 20 years and some of the viruses have resulted in high mortality in humans, a universal influenza vaccine is needed to provide comprehensive protection against a wide range of influenza viruses. Current seasonal influenza vaccines provide strain-specific protection and are less effective against mismatched strains. The rapid antigenic drift and shift in influenza viruses resulted in time-consuming surveillance and uncertainty in the vaccine protection efficacy. Most recent universal influenza vaccine studies target the conserved antigen domains of the viral surface glycoproteins and internal proteins to provide broader protection. Following the development of advanced vaccine technologies, several innovative strategies and vaccine platforms are being explored to generate robust cross-protective immunity. This review provides the latest progress in the development of universal influenza vaccines.
Collapse
Affiliation(s)
- Wen-Chien Wang
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
- Correspondence: (S.S.); (S.K.M.)
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
- Correspondence: (S.S.); (S.K.M.)
| |
Collapse
|
17
|
Gómez-Aguado I, Rodríguez-Castejón J, Beraza-Millor M, Rodríguez-Gascón A, Del Pozo-Rodríguez A, Solinís MÁ. mRNA delivery technologies: Toward clinical translation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:207-293. [PMID: 36064265 DOI: 10.1016/bs.ircmb.2022.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Messenger RNA (mRNA)-therapies have recently taken a huge step toward clinic thanks to the first mRNA-based medicinal products marketed. mRNA features for clinical purposes are improved by chemical modifications, but the inclusion in a delivery system is a regular requirement. mRNA nanomedicines must be designed for the specific therapeutic purpose, protecting the nucleic acid and facilitating the overcoming of biological barriers. Polymers, polypeptides, and cationic lipids are the main used materials to design mRNA delivery systems. Among them, lipid nanoparticles (LNPs) are the most advanced ones, and currently they are at the forefront of preclinical and clinical evaluation in several fields, including immunotherapy (against infectious diseases and cancer), protein replacement, gene editing and regenerative medicine. This chapter includes an overview on mRNA delivery technologies, with special interest in LNPs, and the most recent advances in their clinical application. Liposomes are the mRNA delivery technology with the highest clinical translation among LNPs, whereas the first clinical trial of a therapeutic mRNA formulated in exosomes has been recently approved for protein replacement therapy. The first mRNA products approved by the regulatory agencies worldwide are LNP-based mRNA vaccines against viral infections, specifically against the 2019 coronavirus disease (COVID-19). The clinical translation of mRNA-therapies for cancer is mainly focused on three strategies: anti-cancer vaccination by means of delivering cancer antigens or acting as an adjuvant, mRNA-engineered chimeric antigen receptors (CARs) and T-cell receptors (TCRs), and expression of antibodies and immunomodulators. Cancer immunotherapy and, more recently, COVID-19 vaccines spearhead the advance of mRNA clinical use.
Collapse
Affiliation(s)
- Itziar Gómez-Aguado
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Julen Rodríguez-Castejón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Marina Beraza-Millor
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain.
| |
Collapse
|
18
|
Kloczewiak M, Banks JM, Jin L, Brader ML. A Biopharmaceutical Perspective on Higher-Order Structure and Thermal Stability of mRNA Vaccines. Mol Pharm 2022; 19:2022-2031. [PMID: 35715255 PMCID: PMC9257798 DOI: 10.1021/acs.molpharmaceut.2c00092] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/27/2022]
Abstract
Preservation of the integrity of macromolecular higher-order structure is a tenet central to achieving biologic drug and vaccine product stability toward manufacturing, distribution, storage, handling, and administration. Given that mRNA lipid nanoparticles (mRNA-LNPs) are held together by an intricate ensemble of weak forces, there are some intriguing parallels to biologic drugs, at least at first glance. However, mRNA vaccines are not without unique formulation and stabilization challenges derived from the instability of unmodified mRNA and its limited history as a drug or vaccine. Since certain learning gained from biologic drug development may be applicable for the improvement of mRNA vaccines, we present a perspective on parallels and contrasts between the emerging role of higher-order structure pertaining to mRNA-LNPs compared to pharmaceutical proteins. In a recent publication, the location of mRNA encapsulated within lipid nanoparticles was identified, revealing new insights into the LNP structure, nanoheterogeneity, and microenvironment of the encapsulated mRNA molecules [Brader et al. Biophys. J. 2021, 120, 2766]. We extend those findings by considering the effect of encapsulation on mRNA thermal unfolding with the observation that encapsulation in LNPs increases mRNA unfolding temperatures.
Collapse
Affiliation(s)
- Marek Kloczewiak
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Jessica M. Banks
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Lin Jin
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Mark L. Brader
- Moderna, Inc., 200 Technology Square, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
19
|
Avci-Adali M, A Santos H. Current trends in delivery of non-viral nucleic acid-based therapeutics for improved efficacy. Adv Drug Deliv Rev 2022; 185:114297. [PMID: 35427721 PMCID: PMC9005216 DOI: 10.1016/j.addr.2022.114297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstr. 7/1, 72076 Tuebingen, Germany.
| | - Hélder A Santos
- Head of Department, Department of Biomedical Engineering and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen/University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
20
|
Cui L, Hunter MR, Sonzini S, Pereira S, Romanelli SM, Liu K, Li W, Liang L, Yang B, Mahmoudi N, Desai AS. Mechanistic Studies of an Automated Lipid Nanoparticle Reveal Critical Pharmaceutical Properties Associated with Enhanced mRNA Functional Delivery In Vitro and In Vivo. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105832. [PMID: 34914866 DOI: 10.1002/smll.202105832] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/21/2021] [Indexed: 06/14/2023]
Abstract
Recently, lipid nanoparticles (LNPs) have attracted attention due to their emergent use for COVID-19 mRNA vaccines. The success of LNPs can be attributed to ionizable lipids, which enable functional intracellular delivery. Previously, the authors established an automated high-throughput platform to screen ionizable lipids and identified that the LNPs generated using this automated technique show comparable or increased mRNA functional delivery in vitro as compared to LNPs prepared using traditional microfluidics techniques. In this study, the authors choose one benchmark lipid, DLin-MC3-DMA (MC3), and investigate whether the automated formulation technique can enhance mRNA functional delivery in vivo. Interestingly, a 4.5-fold improvement in mRNA functional delivery in vivo by automated LNPs as compared to LNPs formulated by conventional microfluidics techniques, is observed. Mechanistic studies reveal that particles with large size accommodate more mRNA per LNP, possess more hydrophobic surface, are more hemolytic, bind a larger protein corona, and tend to accumulate more in macropinocytosomes, which may quantitatively benefit mRNA cytosolic delivery. These data suggest that mRNA loading per particle is a critical factor that accounts for the enhanced mRNA functional delivery of automated LNPs. These mechanistic findings provide valuable insight underlying the enhanced mRNA functional delivery to accelerate future mRNA LNP product development.
Collapse
Affiliation(s)
- Lili Cui
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Morag R Hunter
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Silvia Sonzini
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Sara Pereira
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Steven M Romanelli
- University of Michigan Medical School, Department of Molecular & Integrative Physiology, Ann Arbor, MI, 48109-5624, USA
| | - Kai Liu
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43183, Sweden
| | - Weimin Li
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Lihuan Liang
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal & Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Bin Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Najet Mahmoudi
- Rutherford Appleton Laboratory, ISIS Facility, Science and Technology Facilities Council, Didcot, OX11 0QX, UK
| | - Arpan S Desai
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| |
Collapse
|
21
|
Chakritbudsabong W, Sariya L, Jantahiran P, Chaisilp N, Chaiwattanarungruengpaisan S, Rungsiwiwut R, Ferreira JN, Rungarunlert S. Generation of Porcine Induced Neural Stem Cells Using the Sendai Virus. Front Vet Sci 2022; 8:806785. [PMID: 35097051 PMCID: PMC8790232 DOI: 10.3389/fvets.2021.806785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
The reprogramming of cells into induced neural stem cells (iNSCs), which are faster and safer to generate than induced pluripotent stem cells, holds tremendous promise for fundamental and frontier research, as well as personalized cell-based therapies for neurological diseases. However, reprogramming cells with viral vectors increases the risk of tumor development due to vector and transgene integration in the host cell genome. To circumvent this issue, the Sendai virus (SeV) provides an alternative integration-free reprogramming method that removes the danger of genetic alterations and enhances the prospects of iNSCs from bench to bedside. Since pigs are among the most successful large animal models in biomedical research, porcine iNSCs (piNSCs) may serve as a disease model for both veterinary and human medicine. Here, we report the successful generation of piNSC lines from pig fibroblasts by employing the SeV. These piNSCs can be expanded for up to 40 passages in a monolayer culture and produce neurospheres in a suspension culture. These piNSCs express high levels of NSC markers (PAX6, SOX2, NESTIN, and VIMENTIN) and proliferation markers (KI67) using quantitative immunostaining and western blot analysis. Furthermore, piNSCs are multipotent, as they are capable of producing neurons and glia, as demonstrated by their expressions of TUJ1, MAP2, TH, MBP, and GFAP proteins. During the reprogramming of piNSCs with the SeV, no induced pluripotent stem cells developed, and the established piNSCs did not express OCT4, NANOG, and SSEA1. Hence, the use of the SeV can reprogram porcine somatic cells without first going through an intermediate pluripotent state. Our research produced piNSCs using SeV methods in novel, easily accessible large animal cell culture models for evaluating the efficacy of iNSC-based clinical translation in human medicine. Additionally, our piNSCs are potentially applicable in disease modeling in pigs and regenerative therapies in veterinary medicine.
Collapse
Affiliation(s)
- Warunya Chakritbudsabong
- Laboratory of Cellular Biomedicine and Veterinary Medicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Ladawan Sariya
- The Monitoring and Surveillance Center for Zoonotic Disease in Wildlife and Exotic Animals (MoZWE), Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Phakhin Jantahiran
- Laboratory of Cellular Biomedicine and Veterinary Medicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Nattarun Chaisilp
- The Monitoring and Surveillance Center for Zoonotic Disease in Wildlife and Exotic Animals (MoZWE), Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Somjit Chaiwattanarungruengpaisan
- The Monitoring and Surveillance Center for Zoonotic Disease in Wildlife and Exotic Animals (MoZWE), Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Ruttachuk Rungsiwiwut
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | - Joao N. Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Sasitorn Rungarunlert
- Laboratory of Cellular Biomedicine and Veterinary Medicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- *Correspondence: Sasitorn Rungarunlert
| |
Collapse
|
22
|
Mollé LM, Smyth CH, Yuen D, Johnston APR. Nanoparticles for vaccine and gene therapy: Overcoming the barriers to nucleic acid delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1809. [PMID: 36416028 PMCID: PMC9786906 DOI: 10.1002/wnan.1809] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/19/2022] [Accepted: 04/24/2022] [Indexed: 11/24/2022]
Abstract
Nucleic acid therapeutics can be used to control virtually every aspect of cell behavior and therefore have significant potential to treat genetic disorders, infectious diseases, and cancer. However, while clinically approved to treat a small number of diseases, the full potential of nucleic acid therapeutics is hampered by inefficient delivery. Nucleic acids are large, highly charged biomolecules that are sensitive to degradation and so the approaches to deliver these molecules differ significantly from traditional small molecule drugs. Current studies suggest less than 1% of the injected nucleic acid dose is delivered to the target cell in an active form. This inefficient delivery increases costs and limits their use to applications where a small amount of nucleic acid is sufficient. In this review, we focus on two of the major barriers to efficient nucleic acid delivery: (1) delivery to the target cell and (2) transport to the subcellular compartment where the nucleic acids are therapeutically active. We explore how nanoparticles can be modified with targeting ligands to increase accumulation in specific cells, and how the composition of the nanoparticle can be engineered to manipulate or disrupt cellular membranes and facilitate delivery to the optimal subcellular compartments. Finally, we highlight how with intelligent material design, nanoparticle delivery systems have been developed to deliver nucleic acids that silence aberrant genes, correct genetic mutations, and act as both therapeutic and prophylactic vaccines. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Lara M. Mollé
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Cameron H. Smyth
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Daniel Yuen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Angus P. R. Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| |
Collapse
|
23
|
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: 2.0] [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
|
24
|
Ibba ML, Ciccone G, Esposito CL, Catuogno S, Giangrande PH. Advances in mRNA non-viral delivery approaches. Adv Drug Deliv Rev 2021; 177:113930. [PMID: 34403751 DOI: 10.1016/j.addr.2021.113930] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/28/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022]
Abstract
Messenger RNAs (mRNAs) present a great potential as therapeutics for the treatment and prevention of a wide range of human pathologies, allowing for protein replacement, vaccination, cancer immunotherapy, and genomic engineering. Despite advances in the design of mRNA-based therapeutics, a key aspect for their widespread translation to clinic is the development of safe and effective delivery strategies. To this end, non-viral delivery systems including peptide-based complexes, lipidic or polymeric nanoparticles, and hybrid formulations are attracting growing interest. Despite displaying somewhat reduced efficacy compared to viral-based systems, non-viral carriers offer important advantages in terms of biosafety and versatility. In this review, we provide an overview of current mRNA therapeutic applications and discuss key biological barriers to delivery and recent advances in the development of non-viral systems. Challenges and future applications of this novel therapeutic modality are also discussed.
Collapse
Affiliation(s)
- Maria L Ibba
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, 80131 Naples, Italy
| | - Giuseppe Ciccone
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80145 Naples, Italy
| | - Carla L Esposito
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80145 Naples, Italy.
| | - Silvia Catuogno
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80145 Naples, Italy.
| | - Paloma H Giangrande
- University of Iowa, Department of Internal Medicine, Iowa City, IA, USA; Wave Life Sciences, Cambridge, MA, USA.
| |
Collapse
|
25
|
Mayer RL, Impens F. Immunopeptidomics for next-generation bacterial vaccine development. Trends Microbiol 2021; 29:1034-1045. [PMID: 34030969 DOI: 10.1016/j.tim.2021.04.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
Antimicrobial resistance is an increasing global threat and alternative treatments substituting failing antibiotics are urgently needed. Vaccines are recognized as highly effective tools to mitigate antimicrobial resistance; however, the selection of bacterial antigens as vaccine candidates remains challenging. In recent years, advances in mass spectrometry-based proteomics have led to the development of so-called immunopeptidomics approaches that allow the untargeted discovery of bacterial epitopes that are presented on the surface of infected cells. Especially for intracellular bacterial pathogens, immunopeptidomics holds great promise to uncover antigens that can be encoded in viral vector- or nucleic acid-based vaccines. This review provides an overview of immunopeptidomics studies on intracellular bacterial pathogens and considers future directions and challenges in advancing towards next-generation vaccines.
Collapse
Affiliation(s)
- Rupert L Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|
26
|
Rosa SS, Prazeres DMF, Azevedo AM, Marques MPC. mRNA vaccines manufacturing: Challenges and bottlenecks. Vaccine 2021; 39:2190-2200. [PMID: 33771389 PMCID: PMC7987532 DOI: 10.1016/j.vaccine.2021.03.038] [Citation(s) in RCA: 275] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/15/2021] [Accepted: 03/05/2021] [Indexed: 12/20/2022]
Abstract
Vaccines are one of the most important tools in public health and play an important role in infectious diseases control. Owing to its precision, safe profile and flexible manufacturing, mRNA vaccines are reaching the stoplight as a new alternative to conventional vaccines. In fact, mRNA vaccines were the technology of choice for many companies to combat the Covid-19 pandemic, and it was the first technology to be approved in both United States and in Europe Union as a prophylactic treatment. Additionally, mRNA vaccines are being studied in the clinic to treat a number of diseases including cancer, HIV, influenza and even genetic disorders. The increased demand for mRNA vaccines requires a technology platform and cost-effective manufacturing process with a well-defined product characterisation. Large scale production of mRNA vaccines consists in a 1 or 2-step in vitro reaction followed by a purification platform with multiple steps that can include Dnase digestion, precipitation, chromatography or tangential flow filtration. In this review we describe the current state-of-art of mRNA vaccines, focusing on the challenges and bottlenecks of manufacturing that need to be addressed to turn this new vaccination technology into an effective, fast and cost-effective response to emerging health crises.
Collapse
Affiliation(s)
- Sara Sousa Rosa
- Department of Bioengineering, iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Duarte M F Prazeres
- Department of Bioengineering, iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Ana M Azevedo
- Department of Bioengineering, iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Marco P C Marques
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gordon Street, London WC1H 0AH, United Kingdom.
| |
Collapse
|
27
|
Quantum Biotech and Internet of Virus Things: Towards a Theoretical Framework. APPLIED SYSTEM INNOVATION 2021. [DOI: 10.3390/asi4020027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Quantumization, the process of converting information into quantum (qubit) format, is a key enabler for propelling a new and distinct infrastructure in the pharmaceutical space. Quantum messenger RNA (QmRNA) technology, an indispensable constituent of quantum biotech (QB), is a compelling alternative to conventional vaccine methods because of its capacity for rapid development, high efficacy, and low-cost manufacturing to combat infectious diseases. Internet of Virus Things (IoVT), a biological version of Internet of Things (IoT), comprises applications to fight against pandemics and provides effective vaccine administration. The integration of QB and IoVT constitutes the QBIoVT system to advance the prospect of QmRNA vaccine discovery within a few days. This research disseminates the QBIoVT system paradigm, including architectural aspects, priority areas, challenges, applications, and QmRNA research engine design to accelerate QmRNA vaccines discovery. A comprehensive review of the literature was accomplished, and a context-centered methodology was applied to the QBIoVT paradigm forensic investigations to impel QmRNA vaccine discovery. Based on the above rumination, the principal motive for this study was to develop a novel QBIoVT theoretical framework which has not been produced through earlier theories. The proposed framework shall inspire future QBIoVT system research activities to improve pandemics detection and protection.
Collapse
|
28
|
Supabphol S, Li L, Goedegebuure SP, Gillanders WE. Neoantigen vaccine platforms in clinical development: understanding the future of personalized immunotherapy. Expert Opin Investig Drugs 2021; 30:529-541. [PMID: 33641576 DOI: 10.1080/13543784.2021.1896702] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Derived from genetic alterations, cancer neoantigens are proteins with novel amino acid sequences that can be recognized by the immune system. Recent evidence demonstrates that cancer neoantigens represent important targets of cancer immunotherapy. The goal of cancer neoantigen vaccines is to induce neoantigen-specific immune responses and antitumor immunity, while minimizing the potential for autoimmune toxicity. Advances in sequencing technologies, neoantigen prediction ?algorithms,? and other technologies have dramatically improved the ability to identify and prioritize cancer neoantigens. These advances have generated considerable enthusiasm for ?the ?development of neoantigen vaccines. Several neoantigen vaccine platforms are currently being evaluated in early phase clinical trials including the synthetic long peptide (SLP), RNA, dendritic cell (DC), and DNA vaccine platforms. AREAS COVERED In this review, we describe, evaluate the mechanism(s) of action, compare the advantages and disadvantages, and summarize early clinical experience with each vaccine platform. We provide perspectives on the future directions of the neoantigen vaccine field. All data are derived from PubMed and ClinicalTrials search updated in October 2020. EXPERT OPINION Although the initial clinical experience is promising, significant challenges to the success of neoantigen vaccines include limitations in neoantigen identification and the need to successfully target the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Suangson Supabphol
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA.,The Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Lijin Li
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA.,The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA.,The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
29
|
Liu N, Luo Y, Zhu Y, Peng H, Zou C, Zhou Z, Chen W, Wang H, Liu H, Hu Y, Zhang S, Qian K. Effects of Warm Ischemia Time, Cryopreservation, and Grinding Methods on RNA Quality of Mouse Kidney Tissues. Biopreserv Biobank 2021; 19:306-311. [PMID: 33577406 DOI: 10.1089/bio.2020.0129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: High-quality RNA extraction from tissue samples is of key importance for scientific research and translational medicine. Tissue collection and preparation may affect RNA quality. In this study, we investigated effects of warm ischemia time, cryopreservation, and grinding methods on RNA quality. Methods: Total RNA was extracted from mouse kidney tissues with warm ischemia times of 0, 30, 60, 90, and 120 minutes. Half of the tissues were used to extract RNA immediately, while the others were cryopreserved in the vapor phase of liquid nitrogen for 6 months before RNA extraction. A mortar, homogenizer, and tissue lyser were used to grind tissues. RNA was extracted by TRIzol, and RNA integrity was assessed by the RNA integrity number (RIN) value. Results: For fresh tissues and frozen tissues with warm ischemia time within 60 minutes, RIN values were above 7.0 and remained above 6.0 with warm ischemia time within 120 minutes. For the same warm ischemia time, RIN values of frozen tissues were slightly lower than those of fresh tissues. No significant RIN value alterations were observed among grinding methods, but for RNA extraction efficiency, a mortar was much less efficient than the homogenizer or tissue lyser. For frozen tissues, RNA tended to degrade within 8 minutes at room temperature. Conclusions: Mouse kidney tissues with a warm ischemia time within 120 minutes are suitable for general RNA-related research. For tissues with a warm ischemia time within 60 minutes, cryopreservation may not affect RNA quality. The duration of frozen tissues held at room temperature before grinding affects the integrity of RNA, while grinding methods do not affect RNA integrity.
Collapse
Affiliation(s)
- Nan Liu
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Luo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Zhu
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Hongwei Peng
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Cong Zou
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zongning Zhou
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wen Chen
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hui Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huiqin Liu
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Hu
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shanshan Zhang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kaiyu Qian
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| |
Collapse
|
30
|
Mc Cafferty S, De Temmerman J, Kitada T, Becraft JR, Weiss R, Irvine DJ, Devreese M, De Baere S, Combes F, Sanders NN. In Vivo Validation of a Reversible Small Molecule-Based Switch for Synthetic Self-Amplifying mRNA Regulation. Mol Ther 2020; 29:1164-1173. [PMID: 33186690 DOI: 10.1016/j.ymthe.2020.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/03/2020] [Accepted: 11/05/2020] [Indexed: 12/01/2022] Open
Abstract
Synthetic mRNA therapeutics have the potential to revolutionize healthcare, as they enable patients to produce therapeutic proteins inside their own bodies. However, convenient methods that allow external control over the timing and magnitude of protein production after in vivo delivery of synthetic mRNA are lacking. In this study, we validate the in vivo utility of a synthetic self-amplifying mRNA (RNA replicon) whose expression can be turned off using a genetic switch that responds to oral administration of trimethoprim (TMP), a US Food and Drug Administration (FDA)-approved small-molecule drug. After intramuscular electroporation, the engineered RNA replicon exhibited dose-dependent and reversible expression of its encoded protein upon TMP administration. The TMP serum level needed for maximal downregulation of protein translation was approximately 45-fold below that used in humans for therapeutic purposes. To demonstrate the therapeutic potential of the technology, we injected mice with a TMP-responsive RNA replicon encoding erythropoietin (EPO) and successfully controlled the timing and magnitude of EPO production as well as changes in hematocrit. This work demonstrates the feasibility of controlling mRNA kinetics in vivo, thereby broadly expanding the clinical versatility of mRNA therapeutics.
Collapse
Affiliation(s)
- Sean Mc Cafferty
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Joyca De Temmerman
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; Department of Pathology, Bacteriology and Poultry diseases, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | | | | | - Ron Weiss
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Mathias Devreese
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Siegrid De Baere
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
31
|
Younis MA, Khalil IA, Harashima H. Gene Therapy for Hepatocellular Carcinoma: Highlighting the Journey from Theory to Clinical Applications. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mahmoud A. Younis
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Ikramy A. Khalil
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
| |
Collapse
|
32
|
Chen Y, Aslanoglou S, Murayama T, Gervinskas G, Fitzgerald LI, Sriram S, Tian J, Johnston APR, Morikawa Y, Suu K, Elnathan R, Voelcker NH. Silicon-Nanotube-Mediated Intracellular Delivery Enables Ex Vivo Gene Editing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000036. [PMID: 32378244 DOI: 10.1002/adma.202000036] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/16/2020] [Accepted: 03/22/2020] [Indexed: 06/11/2023]
Abstract
Engineered nano-bio cellular interfaces driven by vertical nanostructured materials are set to spur transformative progress in modulating cellular processes and interrogations. In particular, the intracellular delivery-a core concept in fundamental and translational biomedical research-holds great promise for developing novel cell therapies based on gene modification. This study demonstrates the development of a mechanotransfection platform comprising vertically aligned silicon nanotube (VA-SiNT) arrays for ex vivo gene editing. The internal hollow structure of SiNTs allows effective loading of various biomolecule cargoes; and SiNTs mediate delivery of those cargoes into GPE86 mouse embryonic fibroblasts without compromising their viability. Focused ion beam scanning electron microscopy (FIB-SEM) and confocal microscopy results demonstrate localized membrane invaginations and accumulation of caveolin-1 at the cell-NT interface, suggesting the presence of endocytic pits. Small-molecule inhibition of endocytosis suggests that active endocytic process plays a role in the intracellular delivery of cargo from SiNTs. SiNT-mediated siRNA intracellular delivery shows the capacity to reduce expression levels of F-actin binding protein (Triobp) and alter the cellular morphology of GPE86. Finally, the successful delivery of Cas9 ribonucleoprotein (RNP) to specifically target mouse Hprt gene is achieved. This NT-enhanced molecular delivery platform has strong potential to support gene editing technologies.
Collapse
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
| | - Stella Aslanoglou
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Gediminas Gervinskas
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, 15 Innovation Walk, Clayton, VIC, 3800, Australia
| | - Laura I Fitzgerald
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Sharath Sriram
- MicroNano Research Facility (MNRF), RMIT University, Melbourne City Campus, Building 7, Level 4, Bowen Street, Melbourne, VIC, 3000, Australia
| | - Jie Tian
- MicroNano Research Facility (MNRF), RMIT University, Melbourne City Campus, Building 7, Level 4, Bowen Street, Melbourne, VIC, 3000, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Department of Materials Science and Engineering Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
- Department of Materials Science and Engineering Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
- INM-Leibniz Institute for New Materials, Campus D2 2, Saarbrücken, 66123, Germany
| |
Collapse
|
33
|
Forterre AV, Wang JH, Delcayre A, Kim K, Green C, Pegram MD, Jeffrey SS, Matin AC. Extracellular Vesicle-Mediated In Vitro Transcribed mRNA Delivery for Treatment of HER2 + Breast Cancer Xenografts in Mice by Prodrug CB1954 without General Toxicity. Mol Cancer Ther 2020; 19:858-867. [PMID: 31941722 PMCID: PMC7056535 DOI: 10.1158/1535-7163.mct-19-0928] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/02/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023]
Abstract
Prodrugs are harmless until activated by a bacterial or viral gene product; they constitute the basis of gene-delivered prodrug therapies called GDEPT, which can kill tumors without major side effects. Previously, we utilized the prodrug CNOB (C16H7CIN2O4; not clinically tested) and enzyme HChrR6 in GDEPT to generate the drug MCHB (C16H9CIN2O2) in tumors. Extracellular vesicles (EVs) were used for directed gene delivery and HChrR6 mRNA as gene. Here, the clinical transfer of this approach is enhanced by: (i) use of CB1954 (tretazicar) for which safe human dose is established; HChrR6 can activate this prodrug. (ii) EVs delivered in vitro transcribed (IVT) HChrR6 mRNA, eliminating the potentially harmful plasmid transfection of EV producer cells we utilized previously; this has not been done before. IVT mRNA loading of EVs required several steps. Naked mRNA being unstable, we ensured its prodrug activating functionality at each step. This was not possible using tretazicar itself; we relied instead on HChrR6's ability to convert CNOB into MCHB, whose fluorescence is easily visualizable. HChrR6 mRNA-translated product's ability to generate fluorescence from CNOB vicariously indicated its competence for tretazicar activation. (iii) Systemic IVT mRNA-loaded EVs displaying an anti-HER2 single-chain variable fragment ("IVT EXO-DEPTs") and tretazicar caused growth arrest of human HER2+ breast cancer xenografts in athymic mice. As this occurred without injury to other tissues, absence of off-target mRNA delivery is strongly indicated. Many cancer sites are not amenable for direct gene injection, but current GDEPTs require this. In circumventing this need, a major advance in GDEPT applicability has been accomplished.
Collapse
Affiliation(s)
- Alexis V Forterre
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California
| | - Jing-Hung Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California
| | | | - Kyuri Kim
- SRI International, Menlo Park, California
| | | | - Mark D Pegram
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - A C Matin
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
34
|
Nanomedicines to Deliver mRNA: State of the Art and Future Perspectives. NANOMATERIALS 2020; 10:nano10020364. [PMID: 32093140 PMCID: PMC7075285 DOI: 10.3390/nano10020364] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/12/2022]
Abstract
The use of messenger RNA (mRNA) in gene therapy is increasing in recent years, due to its unique features compared to plasmid DNA: Transient expression, no need to enter into the nucleus and no risk of insertional mutagenesis. Nevertheless, the clinical application of mRNA as a therapeutic tool is limited by its instability and ability to activate immune responses; hence, mRNA chemical modifications together with the design of suitable vehicles result essential. This manuscript includes a revision of the strategies employed to enhance in vitro transcribed (IVT) mRNA functionality and efficacy, including the optimization of its stability and translational efficiency, as well as the regulation of its immunostimulatory properties. An overview of the nanosystems designed to protect the mRNA and to overcome the intra and extracellular barriers for successful delivery is also included. Finally, the present and future applications of mRNA nanomedicines for immunization against infectious diseases and cancer, protein replacement, gene editing, and regenerative medicine are highlighted.
Collapse
|
35
|
Abstract
Chimeric antigen receptor (CAR) cancer immunotherapy uses autologous immune system's cells, genetically modified, to reinforce the immune system against cancer cells. Genetic modification is usually mediated via viral transfection, despite the risk of insertional oncogenesis and off target side effects. In vitro-transcribed (IVT)-mRNA-mediated transfection could contribute to a much safer CAR therapy, since IVT-mRNA leaves no ultimate genetic residue in recipient cells. In this chapter, the IVT-mRNA generation procedure is described, from the selection of the target of the CAR T-cells, the cloning of the template for the in vitro transcription and the development of several chemical modifications for optimizing the structure and thus the stability of the produced CAR IVT-mRNA molecules. Among various transfection methods to efficiently express the CAR molecule on T-cells' surface, the electroporation and the cationic-lipid mediated transfection of the CAR IVT-mRNAs are described.
Collapse
Affiliation(s)
- Androulla N Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Lefkothea C Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece.
| |
Collapse
|
36
|
Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat Commun 2019; 10:3974. [PMID: 31481662 PMCID: PMC6722139 DOI: 10.1038/s41467-019-11911-5] [Citation(s) in RCA: 316] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 08/09/2019] [Indexed: 12/16/2022] Open
Abstract
Tumor-associated macrophages (TAMs) usually express an M2 phenotype, which enables them to perform immunosuppressive and tumor-promoting functions. Reprogramming these TAMs toward an M1 phenotype could thwart their pro-cancer activities and unleash anti-tumor immunity, but efforts to accomplish this are nonspecific and elicit systemic inflammation. Here we describe a targeted nanocarrier that can deliver in vitro-transcribed mRNA encoding M1-polarizing transcription factors to reprogram TAMs without causing systemic toxicity. We demonstrate in models of ovarian cancer, melanoma, and glioblastoma that infusions of nanoparticles formulated with mRNAs encoding interferon regulatory factor 5 in combination with its activating kinase IKKβ reverse the immunosuppressive, tumor-supporting state of TAMs and reprogram them to a phenotype that induces anti-tumor immunity and promotes tumor regression. We further establish that these nanoreagents are safe for repeated dosing. Implemented in the clinic, this immunotherapy could enable physicians to obviate suppressive tumors while avoiding systemic treatments that disrupt immune homeostasis.
Collapse
|
37
|
Nakajima M, Yoshimatsu S, Sato T, Nakamura M, Okahara J, Sasaki E, Shiozawa S, Okano H. Establishment of induced pluripotent stem cells from common marmoset fibroblasts by RNA-based reprogramming. Biochem Biophys Res Commun 2019; 515:593-599. [DOI: 10.1016/j.bbrc.2019.05.175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/30/2019] [Indexed: 10/26/2022]
|
38
|
Steinle H, Weber M, Behring A, Mau-Holzmann U, von Ohle C, Popov AF, Schlensak C, Wendel HP, Avci-Adali M. Reprogramming of Urine-Derived Renal Epithelial Cells into iPSCs Using srRNA and Consecutive Differentiation into Beating Cardiomyocytes. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:907-921. [PMID: 31476669 PMCID: PMC6723182 DOI: 10.1016/j.omtn.2019.07.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/26/2019] [Accepted: 07/22/2019] [Indexed: 12/13/2022]
Abstract
The generation of induced pluripotent stem cells (iPSCs) from patient’s somatic cells and the subsequent differentiation into desired cell types opens up numerous possibilities in regenerative medicine and tissue engineering. Adult cardiomyocytes have limited self-renewal capacity; thus, the efficient, safe, and clinically applicable generation of autologous cardiomyocytes is of great interest for the treatment of damaged myocardium. In this study, footprint-free iPSCs were successfully generated from urine-derived renal epithelial cells through a single application of self-replicating RNA (srRNA). The expression of pluripotency markers and the in vitro as well as in vivo trilineage differentiation were demonstrated. Furthermore, the resulting iPSCs contained no residual srRNA, and the karyotyping analysis demonstrated no detectable anomalies. The cardiac differentiation of these iPSCs resulted in autologous contracting cardiomyocytes after 10 days. We anticipate that the use of urine as a non-invasive cell source to obtain patient cells and the use of srRNA for reprogramming into iPSCs will greatly improve the future production of clinically applicable cardiomyocytes and other cell types. This could allow the regeneration of tissues by generating sufficient quantities of autologous cells without the risk of immune rejection.
Collapse
Affiliation(s)
- Heidrun Steinle
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Marbod Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Andreas Behring
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Ulrike Mau-Holzmann
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Christiane von Ohle
- Department of Conservative Dentistry and Periodontology, Centre of Dentistry, Oral Medicine and Maxillofacial Surgery, University Hospital Tübingen, Osianderstraße 2-8, 72076 Tübingen, Germany
| | - Aron-Frederik Popov
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Hans Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany.
| |
Collapse
|
39
|
Hashiro S, Mitsuhashi M, Yasueda H. Overexpression system for recombinant RNA in Corynebacterium glutamicum using a strong promoter derived from corynephage BFK20. J Biosci Bioeng 2019; 128:255-263. [PMID: 31076339 DOI: 10.1016/j.jbiosc.2019.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/20/2019] [Accepted: 03/07/2019] [Indexed: 01/16/2023]
Abstract
In recent years, it has been shown that recombinant RNA molecules have a great potential in mRNA therapy and as novel agricultural pesticides. We developed a fundamental system for efficient production of target RNA molecules in Corynebacterium glutamicum, composed of a strong promoter named F1 and a terminator derived from corynephage BFK20 in a high-copy number plasmid vector. As a target model RNA for overexpression, we designed and used an RNA molecule [designated U1A*-RNA, ∼160 nucleotides (nt) long] containing a stem/loop II (SL-II, hairpin-II) structure from U1 small nuclear RNA (snRNA), which binds to U1A protein, forming a U1 sn-ribonucleoprotein, which is essential in the pre-mRNA splicing process. C. glutamicum strains harboring the U1A*-RNA expression plasmid were cultured and the total RNA was analyzed. We observed prominent expression of RNA corresponding to the U1A*-RNA transcript along with lower expression of a 3'-region-truncated form of the transcript (∼110 nt) in an rnc (encoding RNase III)-deficient strain. We also found that the produced U1A*-RNA bound to the U1A RNA-binding domain protein, which was separately prepared with C. glutamicum. In a batch cultivation using a fermentor, the total accumulated amount of the target RNA reached about 300 mg/L by 24 h. Thus, our results indicated that our system can serve as an efficient platform for large-scale preparation of an RNA of interest.
Collapse
Affiliation(s)
- Shuhei Hashiro
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Mayu Mitsuhashi
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Hisashi Yasueda
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan; Research and Development Center for Precision Medicine, University of Tsukuba, 1-2 Kasuga, Tsukuba-shi, Ibaraki 305-8550, Japan.
| |
Collapse
|
40
|
Tendulkar G, Ehnert S, Sreekumar V, Chen T, Kaps HP, Golombek S, Wendel HP, Nüssler AK, Avci-Adali M. Exogenous Delivery of Link N mRNA into Chondrocytes and MSCs-The Potential Role in Increasing Anabolic Response. Int J Mol Sci 2019; 20:1716. [PMID: 30959917 PMCID: PMC6479841 DOI: 10.3390/ijms20071716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 12/25/2022] Open
Abstract
Musculoskeletal disorders, such as osteoarthritis and intervertebral disc degeneration are causes of morbidity, which concomitantly burdens the health and social care systems worldwide, with massive costs. Link N peptide has recently been described as a novel anabolic stimulator for intervertebral disc repair. In this study, we analyzed the influence on anabolic response, by delivering synthetic Link N encoding mRNA into primary human chondrocytes and mesenchymal stromal cells (SCP1 cells), Furthermore, both cell types were seeded on knitted titanium scaffolds, and the influence of Link N peptide mRNA for possible tissue engineering applications was investigated. Synthetic modified Link N mRNA was efficiently delivered into both cell types and cell transfection resulted in an enhanced expression of aggrecan, Sox 9, and type II collagen with a decreased expression of type X collagen. Interestingly, despite increased expression of BMP2 and BMP7, BMP signaling was repressed and TGFβ signaling was boosted by Link N transfection in mesenchymal stromal cells, suggesting possible regulatory mechanisms. Thus, the exogenous delivery of Link N peptide mRNA into cells augmented an anabolic response and thereby increased extracellular matrix synthesis. Considering these findings, we suppose that the cultivation of cells on knitted titanium scaffolds and the exogenous delivery of Link N peptide mRNA into cells could mechanically support the stability of tissue-engineered constructs and improve the synthesis of extracellular matrix by seeded cells. This method can provide a potent strategy for articular cartilage and intervertebral disc regeneration.
Collapse
Affiliation(s)
- Gauri Tendulkar
- Siegfried Weller Institute for Trauma Research at the BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany.
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research at the BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany.
| | - Vrinda Sreekumar
- Siegfried Weller Institute for Trauma Research at the BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany.
| | - Tao Chen
- Siegfried Weller Institute for Trauma Research at the BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany.
| | - Hans-Peter Kaps
- Siegfried Weller Institute for Trauma Research at the BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany.
| | - Sonia Golombek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany.
| | - Hans-Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany.
| | - Andreas K Nüssler
- Siegfried Weller Institute for Trauma Research at the BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany.
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany.
| |
Collapse
|
41
|
Lee CH, Ingrole RSJ, Gill HS. Generation of induced pluripotent stem cells using elastin like polypeptides as a non-viral gene delivery system. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165405. [PMID: 30753882 DOI: 10.1016/j.bbadis.2019.01.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 02/07/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have been generated from various somatic cells using different approaches; however, a major restriction of reprogramming methods is the use of viral vectors, which have the risk of causing genome-integration of viral DNA. Here, without a viral vector, we generated iPSCs from mouse fibroblasts using an elastin-like polypeptide (ELP)-based transfection method. Our findings support the possible use of ELPs for delivery of the reprogramming genes in to somatic cells for generation of iPSCs. Results of gel retardation assay demonstrated efficient complexation of ELPs with a plasmid containing the four Yamanaka stem cell factors, Oct-4, Klf4, c-myc, and Sox2. After transfection, the iPSCs showed embryonic stem cell-like characteristics, including expression of endogenous pluripotency genes, differentiation into three germ layer lineages, and formation of teratomas in vivo. Our results demonstrate that ELP-based gene delivery may provide a safe method for use in generation of virus-free and exogenous DNA-free iPSCs, which will be crucial for future applications in stem cell-based therapies.
Collapse
Affiliation(s)
- Chang Hyun Lee
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, United States
| | - Rohan S J Ingrole
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th and Canton, Lubbock, TX 79409, United States.
| |
Collapse
|
42
|
Improving mRNA-Based Therapeutic Gene Delivery by Expression-Augmenting 3' UTRs Identified by Cellular Library Screening. Mol Ther 2018; 27:824-836. [PMID: 30638957 DOI: 10.1016/j.ymthe.2018.12.011] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 01/14/2023] Open
Abstract
Synthetic mRNA has emerged as a powerful tool for the transfer of genetic information, and it is being explored for a variety of therapeutic applications. Many of these applications require prolonged intracellular persistence of mRNA to improve bioavailability of the encoded protein. mRNA molecules are intrinsically unstable and their intracellular kinetics depend on the UTRs embracing the coding sequence, in particular the 3' UTR elements. We describe here a novel and generally applicable cell-based selection process for the identification of 3' UTRs that augment the expression of proteins encoded by synthetic mRNA. Moreover, we show, for two applications of mRNA therapeutics, namely, (1) the delivery of vaccine antigens in order to mount T cell immune responses and (2) the introduction of reprogramming factors into differentiated cells in order to induce pluripotency, that mRNAs tagged with the 3' UTR elements discovered in this study outperform those with commonly used 3' UTRs. This approach further leverages the utility of mRNA as a gene therapy drug format.
Collapse
|
43
|
Xu Y, Huang L, Kirschman JL, Vanover DA, Tiwari PM, Santangelo PJ, Shen X, Russell DG. Exploitation of Synthetic mRNA To Drive Immune Effector Cell Recruitment and Functional Reprogramming In Vivo. THE JOURNAL OF IMMUNOLOGY 2018; 202:608-617. [PMID: 30541883 DOI: 10.4049/jimmunol.1800924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022]
Abstract
Therapeutic strategies based on in vitro-transcribed mRNA (IVT) are attractive because they avoid the permanent signature of genomic integration that is associated with DNA-based therapy and result in the transient production of proteins of interest. To date, IVT has mainly been used in vaccination protocols to generate immune responses to foreign Ags. In this "proof-of-principle" study, we explore a strategy of combinatorial IVT to recruit and reprogram immune effector cells to acquire divergent biological functions in mice in vivo. First, we demonstrate that synthetic mRNA encoding CCL3 is able to recruit murine monocytes in a nonprogrammed state, exhibiting neither bactericidal nor tissue-repairing properties. However, upon addition of either Ifn-γ mRNA or Il-4 mRNA, we successfully polarized these cells to adopt either M1 or M2 macrophage activation phenotypes. This cellular reprogramming was demonstrated through increased expression of known surface markers and through the differential modulation of NADPH oxidase activity, or the superoxide burst. Our study demonstrates how IVT strategies can be combined to recruit and reprogram immune effector cells that have the capacity to fulfill complex biological tasks in vivo.
Collapse
Affiliation(s)
- Yitian Xu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853.,Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Lu Huang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Jonathan L Kirschman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Daryll A Vanover
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Pooja M Tiwari
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Xiling Shen
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853.,School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853; and.,Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - David G Russell
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853;
| |
Collapse
|
44
|
Micheu MM, Scarlatescu AI, Scafa-Udriste A, Dorobantu M. The Winding Road of Cardiac Regeneration-Stem Cell Omics in the Spotlight. Cells 2018; 7:255. [PMID: 30544622 PMCID: PMC6315576 DOI: 10.3390/cells7120255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 12/18/2022] Open
Abstract
Despite significant progress in treating ischemic cardiac disease and succeeding heart failure, there is still an unmet need to develop effective therapeutic strategies given the persistent high-mortality rate. Advances in stem cell biology hold great promise for regenerative medicine, particularly for cardiac regeneration. Various cell types have been used both in preclinical and clinical studies to repair the injured heart, either directly or indirectly. Transplanted cells may act in an autocrine and/or paracrine manner to improve the myocyte survival and migration of remote and/or resident stem cells to the site of injury. Still, the molecular mechanisms regulating cardiac protection and repair are poorly understood. Stem cell fate is directed by multifaceted interactions between genetic, epigenetic, transcriptional, and post-transcriptional mechanisms. Decoding stem cells' "panomic" data would provide a comprehensive picture of the underlying mechanisms, resulting in patient-tailored therapy. This review offers a critical analysis of omics data in relation to stem cell survival and differentiation. Additionally, the emerging role of stem cell-derived exosomes as "cell-free" therapy is debated. Last but not least, we discuss the challenges to retrieve and analyze the huge amount of publicly available omics data.
Collapse
Affiliation(s)
- Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Alina Ioana Scarlatescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Alexandru Scafa-Udriste
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| | - Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| |
Collapse
|
45
|
Cao Y, Chen H, Qiu R, Hanna M, Ma E, Hjort M, Zhang A, Lewis RS, Wu JC, Melosh NA. Universal intracellular biomolecule delivery with precise dosage control. SCIENCE ADVANCES 2018; 4:eaat8131. [PMID: 30402539 PMCID: PMC6209385 DOI: 10.1126/sciadv.aat8131] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/02/2018] [Indexed: 05/06/2023]
Abstract
Intracellular delivery of mRNA, DNA, and other large macromolecules into cells plays an essential role in an array of biological research and clinical therapies. However, current methods yield a wide variation in the amount of material delivered, as well as limitations on the cell types and cargoes possible. Here, we demonstrate quantitatively controlled delivery into a range of primary cells and cell lines with a tight dosage distribution using a nanostraw-electroporation system (NES). In NES, cells are cultured onto track-etched membranes with protruding nanostraws that connect to the fluidic environment beneath the membrane. The tight cell-nanostraw interface focuses applied electric fields to the cell membrane, enabling low-voltage and nondamaging local poration of the cell membrane. Concurrently, the field electrophoretically injects biomolecular cargoes through the nanostraws and into the cell at the same location. We show that the amount of material delivered is precisely controlled by the applied voltage, delivery duration, and reagent concentration. NES is highly effective even for primary cell types or different cell densities, is largely cargo agnostic, and can simultaneously deliver specific ratios of different molecules. Using a simple cell culture well format, the NES delivers into >100,000 cells within 20 s with >95% cell viability, enabling facile, dosage-controlled intracellular delivery for a wide variety of biological applications.
Collapse
Affiliation(s)
- Y. Cao
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - H. Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - R. Qiu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - M. Hanna
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - E. Ma
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - M. Hjort
- Division of Synchrotron Radiation Research and Nanometer Structure Consortium, Lund University, Lund, Sweden
| | - A. Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - R. S. Lewis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - J. C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - N. A. Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
46
|
The potential of human induced pluripotent stem cells for modelling diabetic wound healing in vitro. Clin Sci (Lond) 2018; 132:1629-1643. [PMID: 30108152 DOI: 10.1042/cs20171483] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/28/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022]
Abstract
Impaired wound healing and ulceration caused by diabetes mellitus, is a significant healthcare burden, markedly impairs quality of life for patients, and is the major cause of amputation worldwide. Current experimental approaches used to investigate the complex wound healing process often involve cultures of fibroblasts and/or keratinocytes in vitro, which can be limited in terms of complexity and capacity, or utilisation of rodent models in which the mechanisms of wound repair differ substantively from that in humans. However, advances in tissue engineering, and the discovery of strategies to reprogramme adult somatic cells to pluripotency, has led to the possibility of developing models of human skin on a large scale. Generation of induced pluripotent stem cells (iPSCs) from tissues donated by diabetic patients allows the (epi)genetic background of this disease to be studied, and the ability to differentiate iPSCs to multiple cell types found within skin may facilitate the development of more complex skin models; these advances offer key opportunities for improving modelling of wound healing in diabetes, and the development of effective therapeutics for treatment of chronic wounds.
Collapse
|
47
|
Kim BE, Choi SW, Shin JH, Kim JJ, Kang I, Lee BC, Lee JY, Kook MG, Kang KS. Single-Factor SOX2 Mediates Direct Neural Reprogramming of Human Mesenchymal Stem Cells via Transfection of In Vitro Transcribed mRNA. Cell Transplant 2018; 27:1154-1167. [PMID: 29909688 PMCID: PMC6158546 DOI: 10.1177/0963689718771885] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 12/28/2022] Open
Abstract
Neural stem cells (NSCs) are a prominent cell source for understanding neural pathogenesis and for developing therapeutic applications to treat neurodegenerative disease because of their regenerative capacity and multipotency. Recently, a variety of cellular reprogramming technologies have been developed to facilitate in vitro generation of NSCs, called induced NSCs (iNSCs). However, the genetic safety aspects of established virus-based reprogramming methods have been considered, and non-integrating reprogramming methods have been developed. Reprogramming with in vitro transcribed (IVT) mRNA is one of the genetically safe reprogramming methods because exogenous mRNA temporally exists in the cell and is not integrated into the chromosome. Here, we successfully generated expandable iNSCs from human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) via transfection with IVT mRNA encoding SOX2 (SOX2 mRNA) with properly optimized conditions. We confirmed that generated human UCB-MSC-derived iNSCs (UM-iNSCs) possess characteristics of NSCs, including multipotency and self-renewal capacity. Additionally, we transfected human dermal fibroblasts (HDFs) with SOX2 mRNA. Compared with human embryonic stem cell-derived NSCs, HDFs transfected with SOX2 mRNA exhibited neural reprogramming with similar morphologies and NSC-enriched mRNA levels, but they showed limited proliferation ability. Our results demonstrated that human UCB-MSCs can be used for direct reprogramming into NSCs through transfection with IVT mRNA encoding a single factor, which provides an integration-free reprogramming tool for future therapeutic application.
Collapse
Affiliation(s)
- Bo-Eun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- College of Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hee Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jae-Jun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- College of Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Insung Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Byung-Chul Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jin Young Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Myoung Geun Kook
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Patton C, Farr GH, An D, Martini PG, Maves L. Lipid Nanoparticle Packaging Is an Effective and Nontoxic mRNA Delivery Platform in Embryonic Zebrafish. Zebrafish 2018; 15:217-227. [DOI: 10.1089/zeb.2017.1511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Clay Patton
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Gist H. Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Ding An
- Moderna Therapeutics, Cambridge, Massachusetts
| | | | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
| |
Collapse
|
49
|
Lescan M, Perl RM, Golombek S, Pilz M, Hann L, Yasmin M, Behring A, Keller T, Nolte A, Gruhn F, Kochba E, Levin Y, Schlensak C, Wendel HP, Avci-Adali M. De Novo Synthesis of Elastin by Exogenous Delivery of Synthetic Modified mRNA into Skin and Elastin-Deficient Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:475-484. [PMID: 29858082 PMCID: PMC5992474 DOI: 10.1016/j.omtn.2018.03.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 03/05/2018] [Accepted: 03/27/2018] [Indexed: 12/11/2022]
Abstract
Elastin is one of the most important and abundant extracellular matrix (ECM) proteins that provide elasticity and resilience to tissues and organs, including vascular walls, ligaments, skin, and lung. Besides hereditary diseases, such as Williams-Beuren syndrome (WBS), which results in reduced elastin synthesis, injuries, aging, or acquired diseases can lead to the degradation of existing elastin fibers. Thus, the de novo synthesis of elastin is required in several medical conditions to restore the elasticity of affected tissues. Here, we applied synthetic modified mRNA encoding tropoelastin (TE) for the de novo synthesis of elastin and determined the mRNA-mediated elastin synthesis in cells, as well as ex vivo in porcine skin. EA.hy926 cells, human fibroblasts, and mesenchymal stem cells (MSCs) isolated from a patient with WBS were transfected with 2.5 μg TE mRNA. After 24 hr, the production of elastin was analyzed by Fastin assay and dot blot analyses. Compared with untreated cells, significantly enhanced elastin amounts were detected in TE mRNA transfected cells. The delivered synthetic TE mRNA was even able to significantly increase the elastin production in elastin-deficient MSCs. In porcine skin, approximately 20% higher elastin amount was detected after the intradermal delivery of synthetic mRNA by microinjection. In this study, we demonstrated the successful applicability of synthetic TE encoding mRNA to produce elastin in elastin-deficient cells as well as in skin. Thus, this auspicious mRNA-based integration-free method has a huge potential in the field of regenerative medicine to induce de novo elastin synthesis, e.g., in skin, blood vessels, or alveoli.
Collapse
Affiliation(s)
- Mario Lescan
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Regine Mariette Perl
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Sonia Golombek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Martin Pilz
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Ludmilla Hann
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Mahua Yasmin
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Andreas Behring
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Timea Keller
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Andrea Nolte
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Franziska Gruhn
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Efrat Kochba
- NanoPass Technologies, Ltd., 3 Golda Meir, 7403648 Nes Ziona, Israel
| | - Yotam Levin
- NanoPass Technologies, Ltd., 3 Golda Meir, 7403648 Nes Ziona, Israel
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Hans Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany.
| |
Collapse
|
50
|
Golombek S, Pilz M, Steinle H, Kochba E, Levin Y, Lunter D, Schlensak C, Wendel HP, Avci-Adali M. Intradermal Delivery of Synthetic mRNA Using Hollow Microneedles for Efficient and Rapid Production of Exogenous Proteins in Skin. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:382-392. [PMID: 29858073 PMCID: PMC5992458 DOI: 10.1016/j.omtn.2018.03.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/21/2022]
Abstract
In recent years, synthetic mRNA-based applications to produce desired exogenous proteins in cells have been gaining importance. However, systemic delivery of synthetic mRNA can result in unspecific uptake into undesired cells or organs and, thereby, fail to target desired cells. Thus, local and targeted delivery of synthetic mRNA becomes increasingly important to reach the desired cell types and tissues. In this study, intradermal delivery of synthetic mRNA using a hollow microneedle injection-based method was evaluated. Furthermore, an ex vivo porcine skin model was established to analyze synthetic mRNA-mediated protein expression in the skin following intradermal delivery. Using this model, highly efficient delivery of synthetic mRNA was demonstrated, which resulted in detection of high levels of secretable humanized Gaussia luciferase (hGLuc) protein encoded by the microinjected synthetic mRNA. Interestingly, synthetic mRNA injected without transfection reagent was also able to enter the cells and resulted in protein expression. The established ex vivo porcine skin model can be used to evaluate the successful production of desired proteins after intradermal delivery of synthetic mRNAs before starting with in vivo experiments. Furthermore, the use of microneedles enables patient-friendly, painless, and efficient delivery of synthetic mRNAs into the dermis; thus, this method could be applied for local treatment of different skin diseases as well as for vaccination and immunotherapy.
Collapse
Affiliation(s)
- Sonia Golombek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Martin Pilz
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Heidrun Steinle
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Efrat Kochba
- NanoPass Technologies Ltd., 3 Golda Meir, 7403648 Nes Ziona, Israel
| | - Yotam Levin
- NanoPass Technologies Ltd., 3 Golda Meir, 7403648 Nes Ziona, Israel
| | - Dominique Lunter
- Department of Pharmaceutical Technology, Eberhard Karls University, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Hans Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Calwerstraße 7/1, 72076 Tübingen, Germany.
| |
Collapse
|