1
|
Zhang C, Teng Y, Bai X, Tang M, Stewart W, Chen JJ, Xu X, Zhang XQ. Prevent and Reverse Metabolic Dysfunction-Associated Steatohepatitis and Hepatic Fibrosis via mRNA-Mediated Liver-Specific Antibody Therapy. ACS NANO 2024; 18:34375-34390. [PMID: 39639502 DOI: 10.1021/acsnano.4c13404] [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: 12/07/2024]
Abstract
Chronic exposure of the liver to multiple insults culminates in the development of metabolic dysfunction-associated steatohepatitis (MASH), a complicated metabolic syndrome characterized by hepatic steatosis and inflammation, typically accompanied by progressive fibrosis. Despite extensive clinical evaluation, there remain challenges in MASH drug development, which are primarily due to unsatisfactory efficacy and limited specificity. Strategies to address the unmet medical need for MASH with fibrosis before it reaches the irreversible stage of decompensated cirrhosis are critically needed. Herein, we developed an mRNA-mediated liver-specific antibody therapy for MASH and hepatic fibrosis using a targeted lipid nanoparticle (LNP) delivery system. When encapsulated with IL-11 single-chain variable fragment (scFv)-encoded mRNA, the targeted AA3G LNP (termed mIL11-scFv@AA3G) specifically accumulated in the liver and secreted IL-11 scFv to neutralize overexpressed IL-11 in hepatic environments, thus inhibiting the IL-11 signaling pathway in hepatocytes and hepatic stellate cells. As a preventative regimen, systemic administration of mIL11-scFv@AA3G reversed MASH and prevented the progression to fibrosis in a murine model of early MASH. Notably, mIL11-scFv@AA3G exhibited superior efficacy compared to systemic administration of IL-11 scFv alone, attributed to the sustained antibody expression in the liver, which lasted 18-fold longer than that of IL-11 scFv. When tested in the MASH model with fibrosis, mIL11-scFv@AA3G effectively ameliorated steatosis and resolved fibrosis and inflammation. These findings present a versatile LNP platform targeting liver cell subtypes for the sustained expression of therapeutic antibodies to treat MASH and fibrosis. The developed mRNA-mediated liver-specific antibody therapy offers a promising approach for addressing MASH and holds the potential for expansion to various other diseases.
Collapse
Affiliation(s)
- Chenshuang Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Yilong Teng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Xin Bai
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Maoping Tang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - William Stewart
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Jake Jinkun Chen
- Division of Oral Biology, School of Dental Medicine and Tufts University, Boston, Massachusetts 02111, United States
- Department of Genetics, Molecular and Cell Biology, School of Medicine, Tufts University, Boston, Massachusetts 02111, United States
| | - Xiaoyang Xu
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Xue-Qing Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| |
Collapse
|
2
|
Ganesan A, Arunagiri T, Mani S, Kumaran VR, Sk G, Elumalai S, Kannaiah KP, Chanduluru HK. Mpox treatment evolution: past milestones, present advances, and future directions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03385-0. [PMID: 39225831 DOI: 10.1007/s00210-024-03385-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
An underestimated worldwide health concern, Monkeypox (Mpox) is becoming a bigger menace to the world's population. After smallpox was eradicated in 1970, Mpox was found in a rural region of Africa and quickly spread to other African countries. The etiological agent of the Mpox infection, the Mpox virus, is constantly evolving, and its capability for cross-species transmission led to a global outbreak in 2022 which led to several deaths throughout the world. This review aims to showcase the progressive treatment methods and emerging innovations in the diagnostic and prevention strategies for controlling Mpox. The clinical trial data for antiviral drugs were systematically collected and analyzed using statistical tests to determine the most effective antiviral treatment. Emerging viral protein inhibitors that are under investigation for Mpox treatment were also scrutinized in this review. Additionally, modern diagnostic methods, such as the Streamlined CRISPR On Pod Evaluation platform (SCOPE) and graphene quantum rods were reviewed, and the efficacy of mRNA vaccines with traditional smallpox vaccines used for Mpox were compared. The statistical analysis revealed that tecovirimat (TCV) is the most effective antiviral drug among the other evaluated drugs, showing superior efficacy in clinical trials. Similarly, mRNA vaccines offer greater effectiveness compared to conventional smallpox vaccines. Furthermore, emerging nanomedicine and herbal drug candidates were highlighted as potential future treatments for Mpox. The findings underscore the effectiveness of TCV in treating Mpox and highlight significant advancements in preventive treatments. The review also points to innovative approaches in vaccine technology and potential future therapies, including nanomedicine and herbal remedies, which may enhance Mpox management.
Collapse
Affiliation(s)
- Alagammai Ganesan
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Thirumalai Arunagiri
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Suganandhini Mani
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Vamsi Ravi Kumaran
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Gayathrii Sk
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Sandhiya Elumalai
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Kanaka Parvathi Kannaiah
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India.
| | - Hemanth Kumar Chanduluru
- SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
3
|
Hussain A, Wang M, Yu D, Zhang J, Naseer QA, Ullah A, Milon Essola J, Zhang X. Medical and molecular biophysical techniques as substantial tools in the era of mRNA-based vaccine technology. Biomater Sci 2024; 12:4117-4135. [PMID: 39016519 DOI: 10.1039/d4bm00561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The COVID-19 pandemic prompted the advancement of vaccine technology using mRNA delivery into the host cells. Consequently, mRNA-based vaccines have emerged as a practical approach against SARS-CoV-2 owing to their inherent properties, such as cost-effectiveness, rapid manufacturing, and preservation. These features are vital, especially in resource-constrained regions. Nevertheless, the design of mRNA-based vaccines is intricately intertwined with the refinement of biophysical technologies, thereby establishing their high potential. The preparation of mRNA-based vaccines involves a sequence of phases combining medical and molecular biophysical technologies. Furthermore, their efficiency depends on the capability to optimize their positive attributes, thus paving the way for their subsequent preclinical and clinical evaluations. Using biophysical techniques, the characterization of nucleic acids extends from their initial formulation to their cellular internalization abilities and encapsulation in biomolecule complexes, such as lipid nanoparticles (LNPs), for designing mRNA-based LNPs. Furthermore, nanoparticles are subjected to a series of careful screening steps to assess their physical and chemical characteristics before achieving an optimum formulation suitable for preclinical and clinical studies. This review provides a comprehensive understanding of the fundamental role of biophysical techniques in the complex development of mRNA-based vaccines and their role in the recent success during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Abid Hussain
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Maoye Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Dan Yu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Jiahui Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Qais Ahmad Naseer
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Aftab Ullah
- School of Medicine, Huaqiao University, No. 269 Chenghua North Rd., Quanzhou, Fujian 362021, China.
| | - Julien Milon Essola
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Centre for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
4
|
Zhang M, Hussain A, Hu B, Yang H, Li C, Guo S, Han X, Li B, Dai Y, Cao Y, Chi H, Weng Y, Qin CF, Huang Y. Atavistic strategy for the treatment of hyperuricemia via ionizable liposomal mRNA. Nat Commun 2024; 15:6463. [PMID: 39085241 PMCID: PMC11292028 DOI: 10.1038/s41467-024-50752-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Hyperuricemia is associated with an increased risk of gout, hypertension, diabetes, and cardiovascular diseases. Most mammals maintain normal serum uric acid (SUA) via urate oxidase (Uox), an enzyme that metabolizes poorly-soluble UA to highly-soluble allantoin. In contrast, Uox became a pseudogene in humans and apes over the long course of evolution. Here we demonstrate an atavistic strategy for treating hyperuricemia based on endogenous expression of Uox in hepatocytes mediated by mRNA (mUox) loaded with an ionizable lipid nanoparticle termed iLAND. mUox@iLAND allows effective transfection and protein expression in vitro. A single dose of mUox@iLAND lowers SUA levels for several weeks in two female murine models, including a novel long-lasting model, which is also confirmed by metabolomics analysis. Together with the excellent safety profiles observed in vivo, the proposed mRNA agent demonstrates substantial potential for hyperuricemia therapy and the prevention of associated conditions.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Bo Hu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Chunhui Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Shuai Guo
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Xiaofeng Han
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
- Rigerna Therapeutics Co. Ltd., Beijing, China
| | - Bei Li
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Yuhong Cao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, China
| | - Hang Chi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuhua Weng
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
5
|
Muzammil K, Rayyani S, Abbas Sahib A, Gholizadeh O, Naji Sameer H, Jwad Kazem T, Badran Mohammed H, Ghafouri Kalajahi H, Zainul R, Yasamineh S. Recent Advances in Crimean-Congo Hemorrhagic Fever Virus Detection, Treatment, and Vaccination: Overview of Current Status and Challenges. Biol Proced Online 2024; 26:20. [PMID: 38926669 PMCID: PMC11201903 DOI: 10.1186/s12575-024-00244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus, and zoonosis, and affects large regions of Asia, Southwestern and Southeastern Europe, and Africa. CCHFV can produce symptoms, including no specific clinical symptoms, mild to severe clinical symptoms, or deadly infections. Virus isolation attempts, antigen-capture enzyme-linked immunosorbent assay (ELISA), and reverse transcription polymerase chain reaction (RT-PCR) are all possible diagnostic tests for CCHFV. Furthermore, an efficient, quick, and cheap technology, including biosensors, must be designed and developed to detect CCHFV. The goal of this article is to offer an overview of modern laboratory tests available as well as other innovative detection methods such as biosensors for CCHFV, as well as the benefits and limits of the assays. Furthermore, confirmed cases of CCHF are managed with symptomatic assistance and general supportive care. This study examined the various treatment modalities, as well as their respective limitations and developments, including immunotherapy and antivirals. Recent biotechnology advancements and the availability of suitable animal models have accelerated the development of CCHF vaccines by a substantial margin. We examined a range of potential vaccines for CCHF in this research, comprising nucleic acid, viral particles, inactivated, and multi-epitope vaccines, as well as the present obstacles and developments in this field. Thus, the purpose of this review is to present a comprehensive summary of the endeavors dedicated to advancing various diagnostic, therapeutic, and preventive strategies for CCHF infection in anticipation of forthcoming hazards.
Collapse
Affiliation(s)
- Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Khamis Mushait Campus, Abha, 62561, Saudi Arabia
| | - Saba Rayyani
- Medical Faculty, University of Georgi, Tbilisi, Georgia
| | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Tareq Jwad Kazem
- Scientific Affairs Department, Al-Mustaqbal University, Hillah, Babylon, 51001, Iraq
| | - Haneen Badran Mohammed
- Optics techniques department, health and medical techniques college, Al-Noor University, Mosul, Iraq
| | | | - Rahadian Zainul
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Negeri Padang, Padang, Indonesia.
| | - Saman Yasamineh
- Center for Advanced Material Processing, Artificial Intelligence, and Biophysics Informatics (CAMPBIOTICS), Universitas Negeri Padang, Padang, Indonesia.
| |
Collapse
|
6
|
Ye T, Zhou J, Guo C, Zhang K, Wang Y, Liu Y, Zhou J, Xie Y, Li E, Gong R, Zhang J, Chuai X, Chiu S. Polyvalent mpox mRNA vaccines elicit robust immune responses and confer potent protection against vaccinia virus. Cell Rep 2024; 43:114269. [PMID: 38787725 DOI: 10.1016/j.celrep.2024.114269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/14/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The 2022 mpox outbreak led the World Health Organization (WHO) to declare it a public health emergency of international concern (PHEIC). There is a need to develop more effective and safer mpox virus (MPXV)-specific vaccines in response to the mpox epidemic. The mRNA vaccine is a promising platform to protect against MPXV infection. In this study, we construct two bivalent MPXV mRNA vaccines, designated LBA (B6R-A29L) and LAM (A35R-M1R), and a quadrivalent mRNA vaccine, LBAAM (B6R-A35R-A29L-M1R). The immunogenicity and protective efficacy of these vaccines alone or in combination were evaluated in a lethal mouse model. All mRNA vaccine candidates could elicit potential antigen-specific humoral and cellular immune responses and provide protection against vaccinia virus (VACV) infection. The protective effect of the combination of two bivalent mRNA vaccines and the quadrivalent vaccine was superior to that of the individual bivalent mRNA vaccine. Our study provides valuable insights for the development of more efficient and safer mRNA vaccines against mpox.
Collapse
Affiliation(s)
- Tianxi Ye
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinge Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Guo
- Guangzhou Henovcom Bioscience Co., Ltd., Guangzhou, Guangdong 510700, China
| | - Kaiyue Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Yuping Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Yanhui Liu
- Guangzhou Henovcom Bioscience Co., Ltd., Guangzhou, Guangdong 510700, China
| | - Junhui Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yalin Xie
- Guangzhou Henovcom Bioscience Co., Ltd., Guangzhou, Guangdong 510700, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui 230027, China
| | - Rui Gong
- University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China; Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China.
| | - Jiancun Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Science, Guangzhou 510530, China.
| | - Xia Chuai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega Science, Chinese Academy of Sciences, Wuhan, Hubei 430207, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui 230027, China.
| |
Collapse
|
7
|
Li X, Qi J, Wang J, Hu W, Zhou W, Wang Y, Li T. Nanoparticle technology for mRNA: Delivery strategy, clinical application and developmental landscape. Theranostics 2024; 14:738-760. [PMID: 38169577 PMCID: PMC10758055 DOI: 10.7150/thno.84291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
The mRNA vaccine, a groundbreaking advancement in the field of immunology, has garnered international recognition by being awarded the prestigious Nobel Prize, which has emerged as a promising prophylactic and therapeutic modality for various diseases, especially in cancer, rare disease, and infectious disease such as COVID-19, wherein successful mRNA treatment can be achieved by improving the stability of mRNA and introducing a safe and effective delivery system. Nanotechnology-based delivery systems, such as lipid nanoparticles, lipoplexes, polyplexes, lipid-polymer hybrid nanoparticles and others, have attracted great interest and have been explored for mRNA delivery. Nanoscale platforms can protect mRNA from extracellular degradation while promoting endosome escape after endocytosis, hence improving the efficacy. This review provides an overview of diverse nanoplatforms utilized for mRNA delivery in preclinical and clinical stages, including formulation, preparation process, transfection efficiency, and administration route. Furthermore, the market situation and prospects of mRNA vaccines are discussed here.
Collapse
Affiliation(s)
- Xiang Li
- Formulation and Process Development (FPD), WuXi Biologics, 291 Fucheng Road, Hangzhou, 311106, China
| | - Jing Qi
- Formulation and Process Development (FPD), WuXi Biologics, 291 Fucheng Road, Hangzhou, 311106, China
| | - Juan Wang
- Formulation and Process Development (FPD), WuXi Biologics, 291 Fucheng Road, Hangzhou, 311106, China
| | - Weiwei Hu
- WuXi Biologics, 291 Fucheng Road, Hangzhou, 311106, China
| | - Weichang Zhou
- WuXi Biologics, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Yi Wang
- Formulation and Process Development (FPD), WuXi Biologics, 291 Fucheng Road, Hangzhou, 311106, China
| | - Tao Li
- Formulation and Process Development (FPD), WuXi Biologics, 291 Fucheng Road, Hangzhou, 311106, China
| |
Collapse
|
8
|
Liu W, Zhao Y, Fan J, Shen J, Tang H, Tang W, Wu D, Huang W, Ding Y, Qiao P, Lin J, Li Z, Li Q, Cui Q, Liu Y, Chen Y, Pu R, Han X, Yin J, Tan X, Cao G. Smoke and Spike: Benzo[a]pyrene Enhances SARS-CoV-2 Infection by Boosting NR4A2-Induced ACE2 and TMPRSS2 Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300834. [PMID: 37428471 PMCID: PMC10502855 DOI: 10.1002/advs.202300834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/21/2023] [Indexed: 07/11/2023]
Abstract
Cigarette smoke aggravates severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. However, the underlying mechanisms remain unclear. Here, they show that benzo[a]pyrene in cigarette smoke extract facilitates SARS-CoV-2 infection via upregulating angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2). Benzo[a]pyrene trans-activates the promoters of ACE2 and TMPRSS2 by upregulating nuclear receptor subfamily 4 A number 2 (NR4A2) and promoting its binding of NR4A2 to their promoters, which is independent of functional genetic polymorphisms in ACE2 and TMPRSS2. Benzo[a]pyrene increases the susceptibility of lung epithelial cells to SARS-CoV-2 pseudoviruses and facilitates the infection of authentic Omicron BA.5 in primary human alveolar type II cells, lung organoids, and lung and testis of hamsters. Increased expression of Nr4a2, Ace2, and Tmprss2, as well as decreased methylation of CpG islands at the Nr4a2 promoter are observed in aged mice compared to their younger counterparts. NR4A2 knockdown or interferon-λ2/λ3 stimulation downregulates the expression of NR4A2, ACE2, and TMPRSS2, thereby inhibiting the infection. In conclusion, benzo[a]pyrene enhances SARS-CoV-2 infection by boosting NR4A2-induced ACE2 and TMPRSS2 expression. This study elucidates the mechanisms underlying the detrimental effects of cigarette smoking on SARS-CoV-2 infection and provides prophylactic options for coronavirus disease 2019, particularly for the elderly population.
Collapse
|
9
|
Kang DD, Li H, Dong Y. Advancements of in vitro transcribed mRNA (IVT mRNA) to enable translation into the clinics. Adv Drug Deliv Rev 2023; 199:114961. [PMID: 37321375 PMCID: PMC10264168 DOI: 10.1016/j.addr.2023.114961] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
The accelerated progress and approval of two mRNA-based vaccines to address the SARS-CoV-2 virus were unprecedented. This record-setting feat was made possible through the solid foundation of research on in vitro transcribed mRNA (IVT mRNA) which could be utilized as a therapeutic modality. Through decades of thorough research to overcome barriers to implementation, mRNA-based vaccines or therapeutics offer many advantages to rapidly address a broad range of applications including infectious diseases, cancers, and gene editing. Here, we describe the advances that have supported the adoption of IVT mRNA in the clinics, including optimization of the IVT mRNA structural components, synthesis, and lastly concluding with different classes of IVT RNA. Continuing interest in driving IVT mRNA technology will enable a safer and more efficacious therapeutic modality to address emerging and existing diseases.
Collapse
Affiliation(s)
- Diana D Kang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States; Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Haoyuan Li
- Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States; Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center; Dorothy M. Davis Heart & Lung Research Institute, Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, United States; Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
10
|
Chen H, Guo L, Ding J, Zhou W, Qi Y. A General and Efficient Strategy for Gene Delivery Based on Tea Polyphenols Intercalation and Self-Polymerization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302620. [PMID: 37349886 PMCID: PMC10460882 DOI: 10.1002/advs.202302620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/19/2023] [Indexed: 06/24/2023]
Abstract
Gene therapy that employs therapeutic nucleic acids to modulate gene expression has shown great promise for diseases therapy, and its clinical application relies on the development of effective gene vector. Herein a novel gene delivery strategy by just using natural polyphenol (-)-epigallocatechin-3-O-gallate (EGCG) as raw material is reported. EGCG first intercalates into nucleic acids to yield a complex, which then oxidizes and self-polymerizes to form tea polyphenols nanoparticles (TPNs) for effective nucleic acids encapsulation. This is a general method to load any types of nucleic acids with single or double strands and short or long sequences. Such TPNs-based vector achieves comparable gene loading capacity to commonly used cationic materials, but showing lower cytotoxicity. TPNs can effectively penetrate inside cells, escape from endo/lysosomes, and release nucleic acids in response to intracellular glutathione to exert biological functions. To demonstrate the in vivo application, an anti-caspase-3 small interfering ribonucleic acid is loaded into TPNs to treat concanavalin A-induced acute hepatitis, and excellent therapeutic efficacy is obtained in combination with the intrinsic activities of TPNs vector. This work provides a simple, versatile, and cost-effective gene delivery strategy. Given the biocompatibility and intrinsic biofunctions, this TPNs-based gene vector holds great potential to treat various diseases.
Collapse
Affiliation(s)
- Hao Chen
- Department of PathologyZhanjiang Central HospitalGuangdong Medical UniversityZhanjiangGuangdong524000China
- Department of PathologyShihezi University School of MedicineShiheziXinjiang832002China
| | - Lina Guo
- Department of PharmaceuticsXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Jinsong Ding
- Department of PharmaceuticsXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Wenhu Zhou
- Department of PharmaceuticsXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Yan Qi
- Department of PathologyZhanjiang Central HospitalGuangdong Medical UniversityZhanjiangGuangdong524000China
- Department of PathologyShihezi University School of MedicineShiheziXinjiang832002China
| |
Collapse
|
11
|
Yang Z, Li X, Gan X, Wei M, Wang C, Yang G, Zhao Y, Zhu Z, Wang Z. Hydrogel armed with Bmp2 mRNA-enriched exosomes enhances bone regeneration. J Nanobiotechnology 2023; 21:119. [PMID: 37020301 PMCID: PMC10075167 DOI: 10.1186/s12951-023-01871-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Sustained release of bioactive BMP2 (bone morphogenetic protein-2) is important for bone regeneration, while the intrinsic short half-life of BMP2 at protein level cannot meet the clinical need. In this study, we aimed to design Bmp2 mRNA-enriched engineered exosomes, which were then loaded into specific hydrogel to achieve sustained release for more efficient and safe bone regeneration. RESULTS Bmp2 mRNA was enriched into exosomes by selective inhibition of translation in donor cells, in which NoBody (non-annotated P-body dissociating polypeptide, a protein that inhibits mRNA translation) and modified engineered BMP2 plasmids were co-transfected. The derived exosomes were named ExoBMP2+NoBody. In vitro experiments confirmed that ExoBMP2+NoBody had higher abundance of Bmp2 mRNA and thus stronger osteogenic induction capacity. When loaded into GelMA hydrogel via ally-L-glycine modified CP05 linker, the exosomes could be slowly released and thus ensure prolonged effect of BMP2 when endocytosed by the recipient cells. In the in vivo calvarial defect model, ExoBMP2+NoBody-loaded GelMA displayed great capacity in promoting bone regeneration. CONCLUSIONS Together, the proposed ExoBMP2+NoBody-loaded GelMA can provide an efficient and innovative strategy for bone regeneration.
Collapse
Affiliation(s)
- Zhujun Yang
- Department of Stomatology, Xi'an Central Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xuejian Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, Chengdu, China
| | - Mengying Wei
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Chunbao Wang
- College of Chemistry and Bio-Engineering, Yichun University, Yichun, 336000, Jiangxi, China
| | - Guodong Yang
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yimin Zhao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China.
| | - Zhuoli Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, Chengdu, China.
| | - Zhongshan Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
12
|
von der Haar T, Mulroney TE, Hedayioglu F, Kurusamy S, Rust M, Lilley KS, Thaventhiran JE, Willis AE, Smales CM. Translation of in vitro-transcribed RNA therapeutics. Front Mol Biosci 2023; 10:1128067. [PMID: 36845540 PMCID: PMC9943971 DOI: 10.3389/fmolb.2023.1128067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
In vitro transcribed, modified messenger RNAs (IVTmRNAs) have been used to vaccinate billions of individuals against the SARS-CoV-2 virus, and are currently being developed for many additional therapeutic applications. IVTmRNAs must be translated into proteins with therapeutic activity by the same cellular machinery that also translates native endogenous transcripts. However, different genesis pathways and routes of entry into target cells as well as the presence of modified nucleotides mean that the way in which IVTmRNAs engage with the translational machinery, and the efficiency with which they are being translated, differs from native mRNAs. This review summarises our current knowledge of commonalities and differences in translation between IVTmRNAs and cellular mRNAs, which is key for the development of future design strategies that can generate IVTmRNAs with improved activity in therapeutic applications.
Collapse
Affiliation(s)
- Tobias von der Haar
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| | - Thomas E. Mulroney
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - Fabio Hedayioglu
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| | - Sathishkumar Kurusamy
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| | - Maria Rust
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - Kathryn S. Lilley
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - James E. Thaventhiran
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - Anne E. Willis
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, United Kingdom
| | - C. Mark Smales
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
13
|
Vavilis T, Stamoula E, Ainatzoglou A, Sachinidis A, Lamprinou M, Dardalas I, Vizirianakis IS. mRNA in the Context of Protein Replacement Therapy. Pharmaceutics 2023; 15:pharmaceutics15010166. [PMID: 36678793 PMCID: PMC9866414 DOI: 10.3390/pharmaceutics15010166] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Protein replacement therapy is an umbrella term used for medical treatments that aim to substitute or replenish specific protein deficiencies that result either from the protein being absent or non-functional due to mutations in affected patients. Traditionally, such an approach requires a well characterized but arduous and expensive protein production procedure that employs in vitro expression and translation of the pharmaceutical protein in host cells, followed by extensive purification steps. In the wake of the SARS-CoV-2 pandemic, mRNA-based pharmaceuticals were recruited to achieve rapid in vivo production of antigens, proving that the in vivo translation of exogenously administered mRNA is nowadays a viable therapeutic option. In addition, the urgency of the situation and worldwide demand for mRNA-based medicine has led to an evolution in relevant technologies, such as in vitro transcription and nanolipid carriers. In this review, we present preclinical and clinical applications of mRNA as a tool for protein replacement therapy, alongside with information pertaining to the manufacture of modified mRNA through in vitro transcription, carriers employed for its intracellular delivery and critical quality attributes pertaining to the finished product.
Collapse
Affiliation(s)
- Theofanis Vavilis
- Laboratory of Biology and Genetics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Department of Dentistry, European University Cyprus, Nicosia 2404, Cyprus
- Correspondence:
| | - Eleni Stamoula
- Centre of Systems Biology, Department of Biotechnology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Alexandra Ainatzoglou
- Centre of Systems Biology, Department of Biotechnology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athanasios Sachinidis
- 4th Department of Internal Medicine, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Malamatenia Lamprinou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Dardalas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, Nicosia 1700, Cyprus
| |
Collapse
|