1
|
Nie S, Yang B, Ma R, Zha L, Qin Y, Ou L, Chen X, Li L. Synthetic nanomaterials for spleen-specific mRNA delivery. Biomaterials 2025; 314:122859. [PMID: 39362024 DOI: 10.1016/j.biomaterials.2024.122859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
In recent years, mRNA vaccine has achieved increasing interest owing to its high potency, safety, ease of production, and low-cost manufacturing. Currently approved mRNA vaccines are administered intramuscularly to transfect local antigen-presenting cells (APCs) to initiate low to moderate immune responses. Spleen, the largest secondary lymphoid organ in the body which contains a large number of APCs close to B and T lymphocytes, could be the ideal site for effective initiation of an enhanced immune response. Here, we provide an overview of the recent advances in the development of synthetic materials for spleen-specific mRNA delivery, and lipid nanoparticle-based approaches will be highlighted. We further discuss the main challenges for spleen-specific mRNA delivery to provide a reference for the development of next-generation synthetic nanomaterials with optimal properties.
Collapse
Affiliation(s)
- Shihong Nie
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Beiqi Yang
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Ruiying Ma
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China; West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Lili Zha
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yuyang Qin
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Liyuan Ou
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Ling Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
3
|
Wang J, Ding Y, Chong K, Cui M, Cao Z, Tang C, Tian Z, Hu Y, Zhao Y, Jiang S. Recent Advances in Lipid Nanoparticles and Their Safety Concerns for mRNA Delivery. Vaccines (Basel) 2024; 12:1148. [PMID: 39460315 PMCID: PMC11510967 DOI: 10.3390/vaccines12101148] [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: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION The advent of lipid nanoparticles (LNPs) as a delivery platform for mRNA therapeutics has revolutionized the biomedical field, particularly in treating infectious diseases, cancer, genetic disorders, and metabolic diseases. Recent Advances in Therapeutic LNPs: LNPs, composed of ionizable lipids, phospholipids, cholesterol, and polyethylene glycol (PEG) lipids, facilitate efficient cellular uptake and cytosolic release of mRNA while mitigating degradation by nucleases. However, as synthetic entities, LNPs face challenges that alter their therapeutic efficacy and safety concerns. Toxicity/Reactogenicity/Immunogenicity: This review provides a comprehensive overview of the latest advancements in LNP research, focusing on preclinical safety assessments encompassing toxicity, reactogenicity, and immunogenicity. Summary and Outlook: Additionally, it outlines potential strategies for addressing these challenges and offers insights into future research directions for enhancing the application of LNPs in mRNA therapeutics.
Collapse
Affiliation(s)
- Jialiang Wang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yaopeng Ding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kellie Chong
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; (K.C.)
| | - Meng Cui
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Zeyu Cao
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Chenjue Tang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Zhen Tian
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yuping Hu
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; (K.C.)
| | - Yu Zhao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Shaoyi Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
4
|
Cao Q, Fang H, Tian H. mRNA vaccines contribute to innate and adaptive immunity to enhance immune response in vivo. Biomaterials 2024; 310:122628. [PMID: 38820767 DOI: 10.1016/j.biomaterials.2024.122628] [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: 01/06/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Messenger RNA (mRNA) therapeutics have been widely employed as strategies for the treatment and prevention of diseases. Amid the global outbreak of COVID-19, mRNA vaccines have witnessed rapid development. Generally, in the case of mRNA vaccines, the initiation of the innate immune system serves as a prerequisite for triggering subsequent adaptive immune responses. Critical cells, cytokines, and chemokines within the innate immune system play crucial and beneficial roles in coordinating tailored immune reactions towards mRNA vaccines. Furthermore, immunostimulators and delivery systems play a significant role in augmenting the immune potency of mRNA vaccines. In this comprehensive review, we systematically delineate the latest advancements in mRNA vaccine research, present an in-depth exploration of strategies aimed at amplifying the immune effectiveness of mRNA vaccines, and offer some perspectives and recommendations regarding the future advancements in mRNA vaccine development.
Collapse
Affiliation(s)
- Qiannan Cao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China; Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, China; Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China.
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China; Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, China.
| |
Collapse
|
5
|
Jacob EM, Huang J, Chen M. Lipid nanoparticle-based mRNA vaccines: a new frontier in precision oncology. PRECISION CLINICAL MEDICINE 2024; 7:pbae017. [PMID: 39171210 PMCID: PMC11336688 DOI: 10.1093/pcmedi/pbae017] [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: 05/08/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
The delivery of lipid nanoparticle (LNP)-based mRNA therapeutics has captured the attention of the vaccine research community as an innovative and versatile tool for treating a variety of human malignancies. mRNA vaccines are now in the limelight as an alternative to conventional vaccines owing to their high precision, low-cost, rapid manufacture, and superior safety profile. Multiple mRNA vaccine platforms have been developed to target several types of cancer, and many have demonstrated encouraging results in animal models and human trials. The effectiveness of these new mRNA vaccines depends on the efficacy and stability of the antigen(s) of interest generated and the reliability of their delivery to antigen-presenting cells (APCs), especially dendritic cells (DCs). In this review, we provide a detailed overview of mRNA vaccines and their delivery strategies and consider future directions and challenges in advancing and expanding this promising vaccine platform to widespread therapeutic use against cancer.
Collapse
Affiliation(s)
- Eden M Jacob
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| | - Ming Chen
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC 27710, USA
| |
Collapse
|
6
|
Neill B, Romero AR, Fenton OS. Advances in Nonviral mRNA Delivery Materials and Their Application as Vaccines for Melanoma Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4894-4913. [PMID: 37930174 PMCID: PMC11220486 DOI: 10.1021/acsabm.3c00721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Messenger RNA (mRNA) vaccines are promising platforms for cancer immunotherapy because of their potential to encode for a variety of tumor antigens, high tolerability, and capacity to induce strong antitumor immune responses. However, the clinical translation of mRNA cancer vaccines can be hindered by the inefficient delivery of mRNA in vivo. In this review, we provide an overview of mRNA cancer vaccines by discussing their utility in treating melanoma. Specifically, we begin our review by describing the barriers that can impede mRNA delivery to target cells. We then review native mRNA structure and discuss various modification methods shown to enhance mRNA stability and transfection. Next, we outline the advantages and challenges of three nonviral carrier platforms (lipid nanoparticles, polymeric nanoparticles, and lipopolyplexes) frequently used for mRNA delivery. Last, we summarize preclinical and clinical studies that have investigated nonviral mRNA vaccines for the treatment of melanoma. In writing this review, we aim to highlight innovative nonviral strategies designed to address mRNA delivery challenges while emphasizing the exciting potential of mRNA vaccines as next-generation therapies for the treatment of cancers.
Collapse
Affiliation(s)
- Bevin Neill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriana Retamales Romero
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen S. Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
7
|
Long J, Wang Y, Jiang X, Ge J, Chen M, Zheng B, Wang R, Wang M, Xu M, Ke Q, Wang J. Nanomaterials Boost CAR-T Therapy for Solid Tumors. Adv Healthc Mater 2024; 13:e2304615. [PMID: 38483400 DOI: 10.1002/adhm.202304615] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/29/2024] [Indexed: 05/22/2024]
Abstract
T cell engineering, particularly via chimeric antigen receptor (CAR) modifications for enhancing tumor specificity, has shown efficacy in treating hematologic malignancies. The extension of CAR-T cell therapy to solid tumors, however, is impeded by several challenges: The absence of tumor-specific antigens, antigen heterogeneity, a complex immunosuppressive tumor microenvironment, and physical barriers to cell infiltration. Additionally, limitations in CAR-T cell manufacturing capacity and the high costs associated with these therapies restrict their widespread application. The integration of nanomaterials into CAR-T cell production and application offers a promising avenue to mitigate these challenges. Utilizing nanomaterials in the production of CAR-T cells can decrease product variability and lower production expenses, positively impacting the targeting and persistence of CAR-T cells in treatment and minimizing adverse effects. This review comprehensively evaluates the use of various nanomaterials in the production of CAR-T cells, genetic modification, and in vivo delivery. It discusses their underlying mechanisms and potential for clinical application, with a focus on improving specificity and safety in CAR-T cell therapy.
Collapse
Affiliation(s)
- Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, 1001 Xueyuan Road, Shenzhen, 518055, China
| | - Yian Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, 410013, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Junshang Ge
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, 410078, China
| | - Mingfen Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Fujian Medical University, Quanzhou, 362000, China
| | - Boshu Zheng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Rong Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Meifang Xu
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Qi Ke
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| |
Collapse
|
8
|
Hsu JC, Liu P, Song Y, Song W, Saladin RJ, Peng Y, Hu S, Lan X, Cai W. Lymphoid organ-targeted nanomaterials for immunomodulation of cancer, inflammation, and beyond. Chem Soc Rev 2024; 53:7657-7680. [PMID: 38958009 PMCID: PMC11334694 DOI: 10.1039/d4cs00421c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Nanomaterials exhibit significant potential for stimulating immune responses, offering both local and systemic modulation across a variety of diseases. The lymphoid organs, such as the spleen and lymph nodes, are home to various immune cells, including monocytes and dendritic cells, which contribute to both the progression and prevention/treatment of diseases. Consequently, many nanomaterial formulations are being rationally designed to target these organs and engage with specific cell types, thereby inducing therapeutic and protective effects. In this review, we explore crucial cellular interactions and processes involved in immune regulation and highlight innovative nano-based immunomodulatory approaches. We outline essential considerations in nanomaterial design with an emphasis on their impact on biological interactions, targeting capabilities, and treatment efficacy. Through selected examples, we illustrate the strategic targeting of therapeutically active nanomaterials to lymphoid organs and the subsequent immunomodulation for infection resistance, inflammation suppression, self-antigen tolerance, and cancer immunotherapy. Additionally, we address current challenges, discuss emerging topics, and share our outlook on future developments in the field.
Collapse
Affiliation(s)
- Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Peng Liu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Wenyu Song
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Rachel J Saladin
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
9
|
Narasipura EA, Fenton OS. Advances in non-viral mRNA delivery to the spleen. Biomater Sci 2024; 12:3027-3044. [PMID: 38712531 PMCID: PMC11175841 DOI: 10.1039/d4bm00038b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Developing safe and effective delivery strategies for localizing messenger RNA (mRNA) payloads to the spleen is an important goal in the field of genetic medicine. Accomplishing this goal is challenging due to the instability, size, and charge of mRNA payloads. Here, we provide an analysis of non-viral delivery technologies that have been developed to deliver mRNA payloads to the spleen. Specifically, our review begins by outlining the unique anatomy and potential targets for mRNA delivery within the spleen. Next, we describe approaches in mRNA sequence engineering that can be used to improve mRNA delivery to the spleen. Then, we describe advances in non-viral carrier systems that can package and deliver mRNA payloads to the spleen, highlighting key advances in the literature in lipid nanoparticle (LNP) and polymer nanoparticle (PNP) technology platforms. Finally, we provide commentary and outlook on how splenic mRNA delivery may afford next-generation treatments for autoimmune disorders and cancers. In undertaking this approach, our goal with this review is to both establish a fundamental understanding of drug delivery challenges associated with localizing mRNA payloads to the spleen, while also broadly highlighting the potential to use these genetic medicines to treat disease.
Collapse
Affiliation(s)
- Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
10
|
Chu R, Wang Y, Kong J, Pan T, Yang Y, He J. Lipid nanoparticles as the drug carrier for targeted therapy of hepatic disorders. J Mater Chem B 2024; 12:4759-4784. [PMID: 38682294 DOI: 10.1039/d3tb02766j] [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: 05/01/2024]
Abstract
The liver, a complex and vital organ in the human body, is susceptible to various diseases, including metabolic disorders, acute hepatitis, cirrhosis, and hepatocellular carcinoma. In recent decades, these diseases have significantly contributed to global morbidity and mortality. Currently, liver transplantation remains the most effective treatment for hepatic disorders. Nucleic acid therapeutics offer a selective approach to disease treatment through diverse mechanisms, enabling the regulation of relevant genes and providing a novel therapeutic avenue for hepatic disorders. It is expected that nucleic acid drugs will emerge as the third generation of pharmaceuticals, succeeding small molecule drugs and antibody drugs. Lipid nanoparticles (LNPs) represent a crucial technology in the field of drug delivery and constitute a significant advancement in gene therapies. Nucleic acids encapsulated in LNPs are shielded from the degradation of enzymes and effectively delivered to cells, where they are released and regulate specific genes. This paper provides a comprehensive review of the structure, composition, and applications of LNPs in the treatment of hepatic disorders and offers insights into prospects and challenges in the future development of LNPs.
Collapse
Affiliation(s)
- Runxuan Chu
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Yi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Jianglong Kong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Ting Pan
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
- Department of Pharmaceutics School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yani Yang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Jun He
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| |
Collapse
|
11
|
Song J, Zhang Y, Zhou C, Zhan J, Cheng X, Huang H, Mao S, Zong Z. The dawn of a new Era: mRNA vaccines in colorectal cancer immunotherapy. Int Immunopharmacol 2024; 132:112037. [PMID: 38599100 DOI: 10.1016/j.intimp.2024.112037] [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: 02/06/2024] [Revised: 03/24/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
Colorectal cancer (CRC) is a typical cancer that accounts for 10% of all new cancer cases annually and nearly 10% of all cancer deaths. Despite significant progress in current classical interventions for CRC, these traditional strategies could be invasive and with numerous adverse effects. The poor prognosis of CRC patients highlights the evident and pressing need for more efficient and targeted treatment. Novel strategies regarding mRNA vaccines for anti-tumor therapy have also been well-developed since the successful application for the prevention of COVID-19. mRNA vaccine technology won the 2023 Nobel Prize in Physiology or Medicine, signaling a new direction in human anti-cancer treatment: mRNA medicine. As a promising new immunotherapy in CRC and other multiple cancer treatments, the mRNA vaccine has higher specificity, better efficacy, and fewer side effects than traditional strategies. The present review outlines the basics of mRNA vaccines and their advantages over other vaccines and informs an available strategy for developing efficient mRNA vaccines for CRC precise treatment. In the future, more exploration of mRNA vaccines for CRC shall be attached, fostering innovation to address existing limitations.
Collapse
Affiliation(s)
- Jingjing Song
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang 330006, Jiangxi, China; School of Ophthalmology and Optometry, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Yujun Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang 330006, Jiangxi, China; Huankui Academy, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Chulin Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang 330006, Jiangxi, China; The Second Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jianhao Zhan
- Huankui Academy, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Xifu Cheng
- School of Ophthalmology and Optometry, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Haoyu Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang 330006, Jiangxi, China
| | - Shengxun Mao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang 330006, Jiangxi, China.
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
12
|
Alouini S, Pichon C. Therapeutic Vaccines for HPV-Associated Cervical Malignancies: A Systematic Review. Vaccines (Basel) 2024; 12:428. [PMID: 38675811 PMCID: PMC11054545 DOI: 10.3390/vaccines12040428] [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: 02/13/2024] [Revised: 03/07/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
IMPORTANCE Despite widespread prophylactic vaccination, cervical cancer continues to be a major health problem with considerable mortality. Currently, therapeutic vaccines for HPV-associated cervical malignancies are being evaluated as a potential complement to the standard treatment. OBJECTIVE The present systematic review was conducted on randomized controlled trials (RCTs) to investigate the effects of therapeutic vaccines on the treatment of patients with cervical cancer and cervical intraepithelial neoplasia (CIN) of Grades 2 and 3. EVIDENCE REVIEW The PubMed, Embase, and Cochrane Central Register of Controlled Trials databases were searched. Only articles in English published up until 31 January 2024 were selected. Also, reference lists of the selected original papers and recent review articles were manually searched for additional sources. Data on study characteristics were extracted from the selected articles. Data on outcomes of interest were synthesized, and vaccine efficacy endpoints (histological lesion regression, clinical response, and overall survival) were selected as the basis for grouping the studies. FINDINGS After screening 831 articles, nine RCTs with 800 participants were included, of which seven studies with 677 participants involved CIN2 and CIN3 and examined lesion regression to ≤CIN1 as the efficacy endpoint. Results of two of these studies were deemed to have a high risk of bias, and another one did not contain statistical analyses. Results of the other four studies were quantitively synthesized, and the pooling of p-values revealed a significant difference between the vaccine and placebo groups in terms of lesion regression (p-values of 0.135, 0.049, and 0.034 in RCTs, yielding a combined p-value of 0.010). The certainty of the evidence was rated as moderate. Patients with advanced cervical cancers were studied in two RCTs with 123 participants. Clinical response and overall survival were taken as endpoints, and the results were reported as not significant. The certainty of the evidence of these results was rated as very low, mainly due to the very small number of events. All studies reported good tolerance for the vaccines. CONCLUSIONS AND RELEVANCE The results indicate the potential for therapeutic vaccines in the regression of CIN2 and CIN3 lesions. Moreover, a potential gap in evidence is identified regarding the very low number of RCTs in patients with advanced cervical cancer.
Collapse
Affiliation(s)
- Souhail Alouini
- Departement of Gynecological Surgery, Centre Hospitalier Universitaire d’Orléans, 14 Avenue de l’Hôpital, 45100 Orleans, France
- Faculté de Médecine, Université d’Orléans, 45100 Orleans, France
| | - Chantal Pichon
- Institut Universitaire de France, 1 rue Descartes, 75035 Paris, France;
- INSERM ART ARNm, University of Orléans, 45100 Orleans, France
| |
Collapse
|
13
|
Zhou F, Huang L, Li S, Yang W, Chen F, Cai Z, Liu X, Xu W, Lehto V, Lächelt U, Huang R, Shi Y, Lammers T, Tao W, Xu ZP, Wagner E, Xu Z, Yu H. From structural design to delivery: mRNA therapeutics for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20210146. [PMID: 38855617 PMCID: PMC11022630 DOI: 10.1002/exp.20210146] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/15/2023] [Indexed: 06/11/2024]
Abstract
mRNA therapeutics have emerged as powerful tools for cancer immunotherapy in accordance with their superiority in expressing all sequence-known proteins in vivo. In particular, with a small dosage of delivered mRNA, antigen-presenting cells (APCs) can synthesize mutant neo-antigens and multi-antigens and present epitopes to T lymphocytes to elicit antitumor effects. In addition, expressing receptors like chimeric antigen receptor (CAR), T-cell receptor (TCR), CD134, and immune-modulating factors including cytokines, interferons, and antibodies in specific cells can enhance immunological response against tumors. With the maturation of in vitro transcription (IVT) technology, large-scale and pure mRNA encoding specific proteins can be synthesized quickly. However, the clinical translation of mRNA-based anticancer strategies is restricted by delivering mRNA into target organs or cells and the inadequate endosomal escape efficiency of mRNA. Recently, there have been some advances in mRNA-based cancer immunotherapy, which can be roughly classified as modifications of the mRNA structure and the development of delivery systems, especially the lipid nanoparticle platforms. In this review, the latest strategies for overcoming the limitations of mRNA-based cancer immunotherapies and the recent advances in delivering mRNA into specific organs and cells are summarized. Challenges and opportunities for clinical applications of mRNA-based cancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Feng Zhou
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Lujia Huang
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shiqin Li
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Wenfang Yang
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Fangmin Chen
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
| | - Wujun Xu
- Department of Applied PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Vesa‐Pekka Lehto
- Department of Applied PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Ulrich Lächelt
- Department of Pharmaceutical SciencesUniversity of ViennaViennaAustria
| | - Rongqin Huang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug DeliveryMinistry of Education, Fudan UniversityShanghaiChina
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular ImagingRWTH Aachen University ClinicAachenGermany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular ImagingRWTH Aachen University ClinicAachenGermany
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Zhi Ping Xu
- Institute of Biomedical Health Technology and Engineering and Institute of Systems and Physical BiologyShenzhen Bay LaboratoryShenzhenChina
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for NanoscienceLudwig‐Maximilians‐UniversitätMunichGermany
| | - Zhiai Xu
- School of Chemistry and Molecular EngineeringEast China Normal UniversityShanghaiChina
| | - Haijun Yu
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
14
|
Steffens RC, Folda P, Fendler NL, Höhn M, Bücher-Schossau K, Kempter S, Snyder NL, Hartmann L, Wagner E, Berger S. GalNAc- or Mannose-PEG-Functionalized Polyplexes Enable Effective Lectin-Mediated DNA Delivery. Bioconjug Chem 2024; 35:351-370. [PMID: 38440876 DOI: 10.1021/acs.bioconjchem.3c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
A cationic, dendrimer-like oligo(aminoamide) carrier with four-arm topology based on succinoyl tetraethylene pentamine and histidines, cysteines, and N-terminal azido-lysines was screened for plasmid DNA delivery on various cell lines. The incorporated azides allow modification with various shielding agents of different polyethylene glycol (PEG) lengths and/or different ligands by copper-free click reaction, either before or after polyplex formation. Prefunctionalization was found to be advantageous over postfunctionalization in terms of nanoparticle formation, stability, and efficacy. A length of 24 ethylene oxide repetition units and prefunctionalization of ≥50% of azides per carrier promoted optimal polyplex shielding. PEG shielding resulted in drastically reduced DNA transfer, which could be successfully restored by active lectin targeting via novel GalNAc or mannose ligands, enabling enhanced receptor-mediated endocytosis of the carrier system. The involvement of the asialoglycoprotein receptor (ASGPR) in the uptake of GalNAc-functionalized polyplexes was confirmed in the ASGPR-positive hepatocarcinoma cell lines HepG2 and Huh7. Mannose-modified polyplexes showed superior cellular uptake and transfection efficacy compared to unmodified and shielded polyplexes in mannose-receptor-expressing dendritic cell-like DC2.4 cells.
Collapse
Affiliation(s)
- Ricarda C Steffens
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Paul Folda
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Nikole L Fendler
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Katharina Bücher-Schossau
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Susanne Kempter
- Faculty of Physics, LMU Munich, 80539 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Nicole L Snyder
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
- Institute for Macromolecular Chemistry, University Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg im Breisgau, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| |
Collapse
|
15
|
Estapé Senti M, García Del Valle L, Schiffelers RM. mRNA delivery systems for cancer immunotherapy: Lipid nanoparticles and beyond. Adv Drug Deliv Rev 2024; 206:115190. [PMID: 38307296 DOI: 10.1016/j.addr.2024.115190] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
mRNA-based vaccines are emerging as a promising alternative to standard cancer treatments and the conventional vaccines. Moreover, the FDA-approval of three nucleic acid based therapeutics (Onpattro, BNT162b2 and mRNA-1273) has further increased the interest and trust on this type of therapeutics. In order to achieve a significant therapeutic efficacy, the mRNA needs from a drug delivery system. In the last years, several delivery platforms have been explored, being the lipid nanoparticles (LNPs) the most well characterized and studied. A better understanding on how mRNA-based therapeutics operate (both the mRNA itself and the drug delivery system) will help to further improve their efficacy and safety. In this review, we will provide an overview of what mRNA cancer vaccines are and their mode of action and we will highlight the advantages and challenges of the different delivery platforms that are under investigation.
Collapse
Affiliation(s)
- Mariona Estapé Senti
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Lucía García Del Valle
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Raymond M Schiffelers
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
16
|
Imani S, Tagit O, Pichon C. Neoantigen vaccine nanoformulations based on Chemically synthesized minimal mRNA (CmRNA): small molecules, big impact. NPJ Vaccines 2024; 9:14. [PMID: 38238340 PMCID: PMC10796345 DOI: 10.1038/s41541-024-00807-1] [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: 09/03/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Recently, chemically synthesized minimal mRNA (CmRNA) has emerged as a promising alternative to in vitro transcribed mRNA (IVT-mRNA) for cancer therapy and immunotherapy. CmRNA lacking the untranslated regions and polyadenylation exhibits enhanced stability and efficiency. Encapsulation of CmRNA within lipid-polymer hybrid nanoparticles (LPPs) offers an effective approach for personalized neoantigen mRNA vaccines with improved control over tumor growth. LPP-based delivery systems provide superior pharmacokinetics, stability, and lower toxicity compared to viral vectors, naked mRNA, or lipid nanoparticles that are commonly used for mRNA delivery. Precise customization of LPPs in terms of size, surface charge, and composition allows for optimized cellular uptake, target specificity, and immune stimulation. CmRNA-encoded neo-antigens demonstrate high translational efficiency, enabling immune recognition by CD8+ T cells upon processing and presentation. This perspective highlights the potential benefits, challenges, and future directions of CmRNA neoantigen vaccines in cancer therapy compared to Circular RNAs and IVT-mRNA. Further research is needed to optimize vaccine design, delivery, and safety assessment in clinical trials. Nevertheless, personalized LPP-CmRNA vaccines hold great potential for advancing cancer immunotherapy, paving the way for personalized medicine.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Oya Tagit
- Institute of Chemistry and Bioanalytics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | - Chantal Pichon
- Center of Molecular Biophysics, CNRS, Orléans, France.
- ART-ARNm, National Institute of Health and Medical Research (Inserm) and University of Orléans, Orléans, France.
- Institut Universitaire de France, Paris, France.
| |
Collapse
|
17
|
Mochida Y, Uchida S. mRNA vaccine designs for optimal adjuvanticity and delivery. RNA Biol 2024; 21:1-27. [PMID: 38528828 PMCID: PMC10968337 DOI: 10.1080/15476286.2024.2333123] [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] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
Adjuvanticity and delivery are crucial facets of mRNA vaccine design. In modern mRNA vaccines, adjuvant functions are integrated into mRNA vaccine nanoparticles, allowing the co-delivery of antigen mRNA and adjuvants in a unified, all-in-one formulation. In this formulation, many mRNA vaccines utilize the immunostimulating properties of mRNA and vaccine carrier components, including lipids and polymers, as adjuvants. However, careful design is necessary, as excessive adjuvanticity and activation of improper innate immune signalling can conversely hinder vaccination efficacy and trigger adverse effects. mRNA vaccines also require delivery systems to achieve antigen expression in antigen-presenting cells (APCs) within lymphoid organs. Some vaccines directly target APCs in the lymphoid organs, while others rely on APCs migration to the draining lymph nodes after taking up mRNA vaccines. This review explores the current mechanistic understanding of these processes and the ongoing efforts to improve vaccine safety and efficacy based on this understanding.
Collapse
Affiliation(s)
- Yuki Mochida
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Satoshi Uchida
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| |
Collapse
|
18
|
Delehedde C, Ciganek I, Laroui N, Rameix N, Perche F, Pichon C. Messenger RNA Lipid-Based Nanoparticles: Optimization of Formulations in the Lab. Methods Mol Biol 2024; 2786:255-287. [PMID: 38814399 DOI: 10.1007/978-1-0716-3770-8_12] [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] [Indexed: 05/31/2024]
Abstract
Among the large variety of messenger RNA (mRNA) delivery systems, those developed with lipid-based formulations were the most widely used and efficient. In our lab, we produced different mRNA formulations made with liposomes, hybrid lipid polymer, and lipid nanoparticles. Our formulations were made with lipids bearing imidazole groups that trigger the endosomal escape of nanoparticles once protonated inside the mild acidic milieu of endosomes upon their cell uptake. Herein, we describe protocols that we used to produce, optimize, and characterize those formulations. The transfection efficiency is influenced by various factors including the physicochemical parameters of the nanoparticles, their efficiency to be internalized in cells, and their intracellular routing as well as their capacity to induce immune system sensors. We provide details on how to quantify the amount of mRNA nanoparticles uptake by cells and evaluate the acidity of the intracellular compartments where they are located, to investigate the endosomal escape, and to assess the activation of innate immune sensors as phosphorylation of PKR hampering mRNA translation.
Collapse
Affiliation(s)
- Christophe Delehedde
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Ivan Ciganek
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Inserm, ART-ARNm Inserm US55, Orléans, France
| | - Nabila Laroui
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Inserm, ART-ARNm Inserm US55, Orléans, France
| | - Nathalie Rameix
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Federico Perche
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
| | - Chantal Pichon
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France.
- Orléans University, Château de la Source, Orleans, France.
- Institut Universitaire de France, Paris, France.
- Inserm, ART-ARNm Inserm US55, Orléans, France.
| |
Collapse
|
19
|
Zeyn Y, Hobernik D, Wilk U, Pöhmerer J, Hieber C, Medina-Montano C, Röhrig N, Strähle CF, Thoma-Kress AK, Wagner E, Bros M, Berger S. Transcriptional Targeting of Dendritic Cells Using an Optimized Human Fascin1 Gene Promoter. Int J Mol Sci 2023; 24:16938. [PMID: 38069260 PMCID: PMC10706967 DOI: 10.3390/ijms242316938] [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: 10/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Deeper knowledge about the role of the tumor microenvironment (TME) in cancer development and progression has resulted in new strategies such as gene-based cancer immunotherapy. Whereas some approaches focus on the expression of tumoricidal genes within the TME, DNA-based vaccines are intended to be expressed in antigen-presenting cells (e.g., dendritic cells, DCs) in secondary lymphoid organs, which in turn induce anti-tumor T cell responses. Besides effective delivery systems and the requirement of appropriate adjuvants, DNA vaccines themselves need to be optimized regarding efficacy and selectivity. In this work, the concept of DC-focused transcriptional targeting was tested by applying a plasmid encoding for the luciferase reporter gene under the control of a derivative of the human fascin1 gene promoter (pFscnLuc), comprising the proximal core promoter fused to the normally more distantly located DC enhancer region. DC-focused activity of this reporter construct was confirmed in cell culture in comparison to a standard reporter vector encoding for luciferase under the control of the strong ubiquitously active cytomegalovirus promoter and enhancer (pCMVLuc). Both plasmids were also compared upon intravenous administration in mice. The organ- and cell type-specific expression profile of pFscnLuc versus pCMVLuc demonstrated favorable activity especially in the spleen as a central immune organ and within the spleen in DCs.
Collapse
Affiliation(s)
- Yanira Zeyn
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Dominika Hobernik
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Ulrich Wilk
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Jana Pöhmerer
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Christoph Hieber
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Caroline F. Strähle
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.F.S.); (A.K.T.-K.)
| | - Andrea K. Thoma-Kress
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.F.S.); (A.K.T.-K.)
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| |
Collapse
|
20
|
Jeong H, Kim H, Kim YA, Kim KS, Na K. Intranasal mRNA Delivery via Customized RNA-Polyplex Nanoparticles Enhancing Gene Expression through Photochemical Mechanisms. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 38015621 DOI: 10.1021/acsami.3c12604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Achieving effective mRNA expression in vivo requires careful selection of an appropriate delivery vehicle and route of administration. Among the various routes of administration, intranasal administration has received considerable attention due to its ability to induce potent immune responses. In this context, we designed a specialized cationic polymer tailored for delivery of mRNA into the nasal cavity. These polymers are designed with varying degrees of substitution in different amine groups to allow for identification of the most suitable amine moiety for effective mRNA delivery. We also incorporated a photosensitizer within the polymer structure that can trigger the generation of reactive oxygen species when exposed to light. The synthesized cationic polymer is complexed with anionic mRNA to form a polyplex. Illuminating these polyplexes with laser light enhances their escape from intracellular endosomes, stimulating mRNA translocation into the cytoplasm, followed by increased mRNA expression at the cellular level. Through intranasal administration to C57BL/6 mice, it was confirmed that these photoactive polyplexes effectively induce mRNA expression and activate immune responses in vivo using photochemical effects. This innovative design of a photoactivated cationic polymer presents a promising and reliable strategy to achieve efficient intranasal mRNA delivery. This approach has potential applications in the development of mRNA-based vaccines for both prophylactic and therapeutic purposes.
Collapse
Affiliation(s)
- Hayoon Jeong
- Department of Biomedical-Chemical Engineering, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
- Department of Biotechnology, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
| | - Hongjae Kim
- Department of Biomedical-Chemical Engineering, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
- Department of Biotechnology, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
| | - Young A Kim
- Department of Biomedical-Chemical Engineering, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
- Department of Biotechnology, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
| | - Kyoung Sub Kim
- Department of Biotechnology, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
| | - Kun Na
- Department of Biomedical-Chemical Engineering, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
- Department of Biotechnology, the Catholic University of Korea, Jibongro 43, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
| |
Collapse
|
21
|
Youness RA, Mohamed AH, Efthimiadou EK, Mekky RY, Braoudaki M, Fahmy SA. A Snapshot of Photoresponsive Liposomes in Cancer Chemotherapy and Immunotherapy: Opportunities and Challenges. ACS OMEGA 2023; 8:44424-44436. [PMID: 38046305 PMCID: PMC10688172 DOI: 10.1021/acsomega.3c04134] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/18/2023] [Indexed: 12/05/2023]
Abstract
To provide precise medical regimens, photonics technologies have been involved in the field of nanomedicine. Phototriggered liposomes have been cast as promising nanosystems that achieve controlled release of payloads in several pathological conditions such as cancer, autoimmune, and infectious diseases. In contrast to the conventional liposomes, this photoresponsive element greatly improves therapeutic efficacy and reduces the adverse effects of gene/drug therapy during treatment. Recently, cancer immunotherpay has been one of the hot topics in the field of oncology due to the great success and therapeutic benefits that were well-recognized by the patients. However, several side effects have been encountered due to the unmonitored augmentation of the immune system. This Review highlights the most recent advancements in the development of photoresponsive liposome nanosystems in the field of oncology, with a specific emphasis on challenges and opportunities in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Rana A. Youness
- Biology
and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, Cairo 4824201, Egrypt
- Biology
and Biochemistry Department, Molecular Genetics Research Team (MGRT),
School of Life and Medical Sciences, University
of Hertfordshire Hosted by Global Academic Foundation, Cairo 11835, Egypt
| | - Adham H. Mohamed
- Department
of Chemistry, Faculty of Science, Cairo
University, Giza 12613, Egypt
| | - Eleni K. Efthimiadou
- Inorganic
Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou 157 71, Greece
| | - Radwa Y. Mekky
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo 12622, Egypt
| | - Maria Braoudaki
- Clinical,
Pharmaceutical, and Biological Science Department, School of Life
and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, U.K.
| | - Sherif Ashraf Fahmy
- Chemistry
Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo 11835, Egypt
| |
Collapse
|
22
|
Delehedde C, Ciganek I, Rameix N, Laroui N, Gonçalves C, Even L, Midoux P, Pichon C. Impact of net charge, targeting ligand amount and mRNA modification on the uptake, intracellular routing and the transfection efficiency of mRNA lipopolyplexes in dendritic cells. Int J Pharm 2023; 647:123531. [PMID: 37863445 DOI: 10.1016/j.ijpharm.2023.123531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Targeting mRNA formulations to achieve cell specificity is one of the challenges that must be tackled to mettle their therapeutic potential. Here, lipopolyplexes (LPR) bearing tri-mannose-lipid (TM) are used to target mannose receptor on dendritic cells. We investigated the impact of the net charge and percentage of TM units on the binding, uptake, transfection efficiency (TE) and RNA sensors activation. Binding and uptake capacities of naked and targeted LPR increase with the percent of cationic lipid, but the latter are 2-fold more up taken by the cells. Cationic LPR bearing 5 % and 10 % TM were localized in acidic compartments in contrast to naked LPR and 2.5 % TM-LPR. The drawback is the dramatic decrease of TE as the number of TM-units increases. Cationic LPR bearing 5 % and 10 % TM strongly induced NF-κB and PKR phosphorylation at 6 h. Conversely, mTOR is less activated in line with their low TE. Those side effects are overcome by using 5-methoxyuridine mRNA resulting in an improved TE due to non-phosphorylation of NF-κB and PKR and mTOR activation. Our results point out that targeting DC via mannose receptor triggers a higher uptake of cationic LPRs and fast routing to acidic compartments, and that efficient TE requires low number of TM units use or modified mRNA to escape RNA sensors activation to enhance the translation.
Collapse
Affiliation(s)
- Christophe Delehedde
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France; Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Ivan Ciganek
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Nathalie Rameix
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Nabila Laroui
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Cristine Gonçalves
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Luc Even
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, F-45071, Orléans cedex 02, France; Inserm UMS 55 ART ARNm and University of Orléans, F-45100 Orléans; Institut Universitaire de France, 1 rue Descartes, F-75035 Paris, France.
| |
Collapse
|
23
|
Abstract
The use of cancer vaccines is considered a promising therapeutic strategy in clinical oncology, which is achieved by stimulating antitumor immunity with tumor antigens delivered in the form of cells, peptides, viruses, and nucleic acids. The ideal cancer vaccine has many advantages, including low toxicity, specificity, and induction of persistent immune memory to overcome tumor heterogeneity and reverse the immunosuppressive microenvironment. Many therapeutic vaccines have entered clinical trials for a variety of cancers, including melanoma, breast cancer, lung cancer, and others. However, many challenges, including single antigen targeting, weak immunogenicity, off-target effects, and impaired immune response, have hindered their broad clinical translation. In this review, we introduce the principle of action, components (including antigens and adjuvants), and classification (according to applicable objects and preparation methods) of cancer vaccines, summarize the delivery methods of cancer vaccines, and review the clinical and theoretical research progress of cancer vaccines. We also present new insights into cancer vaccine technologies, platforms, and applications as well as an understanding of potential next-generation preventive and therapeutic vaccine technologies, providing a broader perspective for future vaccine design.
Collapse
Affiliation(s)
- Nian Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Xiangyu Xiao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Ziqiang Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
24
|
Liu X, Huang P, Yang R, Deng H. mRNA Cancer Vaccines: Construction and Boosting Strategies. ACS NANO 2023; 17:19550-19580. [PMID: 37819640 DOI: 10.1021/acsnano.3c05635] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
In late 2020, the U.S. Food and Drug Administration (FDA) approved a lipid-based mRNA vaccine for the prevention of COVID-19, which has pushed this field to be more closely studied and motivated researchers to delve deeper into mRNA therapeutics. To date, the research on mRNA cancer vaccines has been developed rapidly, and substantial hopeful therapeutic results have been achieved against various solid tumors in clinical trials. In this review, we first introduce three main components of mRNA cancer vaccines, including mRNA antigens, adjuvants, and delivery vectors. Engineering these components can optimize the therapeutic effects of mRNA cancer vaccines. For instance, appropriate modification of mRNA structure can alleviate the poor stability and innate immunogenicity of mRNA, and the use of mRNA delivery vectors can address the issues of low delivery efficiency in vivo. Second, we emphatically discuss some strategies to further improve the efficacy of mRNA cancer vaccines, namely modulating the immunosuppressive tumor environment, optimizing administration routes, achieving targeting delivery to intended tissues or organs, and employing combination therapy. These strategies can strengthen the tumor inhibitory ability of mRNA cancer vaccines and increase the possibility of tumor elimination. Finally, we point out some challenges in the clinical practice of mRNA cancer vaccines and offer our perspectives on future developments in this rapidly evolving field. It is anticipated that mRNA cancer vaccines will be rapidly developed for clinical cancer therapy in the near future.
Collapse
Affiliation(s)
- Xiaoqing Liu
- School of Advanced Materials and Nanotechnology, Xidian University, Xi'an 710126 China
- Ministry of Education, School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Pei Huang
- Ministry of Education, School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Rusen Yang
- School of Advanced Materials and Nanotechnology, Xidian University, Xi'an 710126 China
| | - Hongzhang Deng
- Ministry of Education, School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Xi'an, Shaanxi 710126, China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| |
Collapse
|
25
|
Lee Y, Jeong M, Park J, Jung H, Lee H. Immunogenicity of lipid nanoparticles and its impact on the efficacy of mRNA vaccines and therapeutics. Exp Mol Med 2023; 55:2085-2096. [PMID: 37779140 PMCID: PMC10618257 DOI: 10.1038/s12276-023-01086-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 10/03/2023] Open
Abstract
Several studies have utilized a lipid nanoparticle delivery system to enhance the effectiveness of mRNA therapeutics and vaccines. However, these nanoparticles are recognized as foreign materials by the body and stimulate innate immunity, which in turn impacts adaptive immunity. Therefore, it is crucial to understand the specific type of innate immune response triggered by lipid nanoparticles. This article provides an overview of the immunological response in the body, explores how lipid nanoparticles activate the innate immune system, and examines the adverse effects and immunogenicity-related development pathways associated with these nanoparticles. Finally, we highlight and explore strategies for regulating the immunogenicity of lipid nanoparticles.
Collapse
Affiliation(s)
- Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Jeongeun Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Hyein Jung
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea.
| |
Collapse
|
26
|
Paris JL, Gaspar R, Coelho F, De Beule PAA, Silva BFB. Stability Criterion for the Assembly of Core-Shell Lipid-Polymer-Nucleic Acid Nanoparticles. ACS NANO 2023; 17:17587-17594. [PMID: 37581895 PMCID: PMC10510699 DOI: 10.1021/acsnano.3c07204] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/16/2023]
Abstract
Hybrid core-shell lipid-polycation-nucleic acid nanoparticles (LPNPs) provide unique delivery strategies for nonviral gene therapeutics. Since LPNPs consist of multiple components, involving different pairwise interactions between them, they are challenging to characterize and understand. Here, we propose a method based on fluorescence cross-correlation spectroscopy to elucidate the association between the three LPNP components. Through this lens, we demonstrate that cationic lipid shells (liposomes) do not displace polycations or DNA from the polycation-DNA cores (polyplexes). Hence, polyplexes and liposomes must be oppositely charged to associate into LPNPs. Furthermore, we identify the liposome:polyplex number ratio (ρN), which was hitherto an intangible quantity, as the primary parameter predicting stable LPNPs. We establish that ρN ≥ 1 ensures that every polyplex is enveloped by a liposome, thus avoiding coexisting oppositely charged species prone to aggregation.
Collapse
Affiliation(s)
| | - Ricardo Gaspar
- International Iberian Nanotechnology
Laboratory, Braga, 4715-330, Portugal
| | - Filipe Coelho
- International Iberian Nanotechnology
Laboratory, Braga, 4715-330, Portugal
| | | | | |
Collapse
|
27
|
Lee DY, Amirthalingam S, Lee C, Rajendran AK, Ahn YH, Hwang NS. Strategies for targeted gene delivery using lipid nanoparticles and cell-derived nanovesicles. NANOSCALE ADVANCES 2023; 5:3834-3856. [PMID: 37496613 PMCID: PMC10368001 DOI: 10.1039/d3na00198a] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/10/2023] [Indexed: 07/28/2023]
Abstract
Gene therapy is a promising approach for the treatment of many diseases. However, the effective delivery of the cargo without degradation in vivo is one of the major hurdles. With the advent of lipid nanoparticles (LNPs) and cell-derived nanovesicles (CDNs), gene delivery holds a very promising future. The targeting of these nanosystems is a prerequisite for effective transfection with minimal side-effects. In this review, we highlight the emerging strategies utilized for the effective targeting of LNPs and CDNs, and we summarize the preparation methodologies for LNPs and CDNs. We have also highlighted the non-ligand targeting of LNPs toward certain organs based on their composition. It is highly expected that continuing the developments in the targeting approaches of LNPs and CDNs for the delivery system will further promote them in clinical translation.
Collapse
Affiliation(s)
- Dong-Yup Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Institute of Engineering Research, Seoul National University Seoul 08826 Republic of Korea
| | - Changyub Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Arun Kumar Rajendran
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Young-Hyun Ahn
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University Seoul 08826 Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University Seoul 08826 Republic of Korea
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University Seoul 08826 Republic of Korea
- Institute of Engineering Research, Seoul National University Seoul 08826 Republic of Korea
| |
Collapse
|
28
|
Chehelgerdi M, Chehelgerdi M. The use of RNA-based treatments in the field of cancer immunotherapy. Mol Cancer 2023; 22:106. [PMID: 37420174 PMCID: PMC10401791 DOI: 10.1186/s12943-023-01807-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 07/09/2023] Open
Abstract
Over the past several decades, mRNA vaccines have evolved from a theoretical concept to a clinical reality. These vaccines offer several advantages over traditional vaccine techniques, including their high potency, rapid development, low-cost manufacturing, and safe administration. However, until recently, concerns over the instability and inefficient distribution of mRNA in vivo have limited their utility. Fortunately, recent technological advancements have mostly resolved these concerns, resulting in the development of numerous mRNA vaccination platforms for infectious diseases and various types of cancer. These platforms have shown promising outcomes in both animal models and humans. This study highlights the potential of mRNA vaccines as a promising alternative approach to conventional vaccine techniques and cancer treatment. This review article aims to provide a thorough and detailed examination of mRNA vaccines, including their mechanisms of action and potential applications in cancer immunotherapy. Additionally, the article will analyze the current state of mRNA vaccine technology and highlight future directions for the development and implementation of this promising vaccine platform as a mainstream therapeutic option. The review will also discuss potential challenges and limitations of mRNA vaccines, such as their stability and in vivo distribution, and suggest ways to overcome these issues. By providing a comprehensive overview and critical analysis of mRNA vaccines, this review aims to contribute to the advancement of this innovative approach to cancer treatment.
Collapse
Affiliation(s)
- Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
29
|
Hieber C, Grabbe S, Bros M. Counteracting Immunosenescence-Which Therapeutic Strategies Are Promising? Biomolecules 2023; 13:1085. [PMID: 37509121 PMCID: PMC10377144 DOI: 10.3390/biom13071085] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Aging attenuates the overall responsiveness of the immune system to eradicate pathogens. The increased production of pro-inflammatory cytokines by innate immune cells under basal conditions, termed inflammaging, contributes to impaired innate immune responsiveness towards pathogen-mediated stimulation and limits antigen-presenting activity. Adaptive immune responses are attenuated as well due to lowered numbers of naïve lymphocytes and their impaired responsiveness towards antigen-specific stimulation. Additionally, the numbers of immunoregulatory cell types, comprising regulatory T cells and myeloid-derived suppressor cells, that inhibit the activity of innate and adaptive immune cells are elevated. This review aims to summarize our knowledge on the cellular and molecular causes of immunosenescence while also taking into account senescence effects that constitute immune evasion mechanisms in the case of chronic viral infections and cancer. For tumor therapy numerous nanoformulated drugs have been developed to overcome poor solubility of compounds and to enable cell-directed delivery in order to restore immune functions, e.g., by addressing dysregulated signaling pathways. Further, nanovaccines which efficiently address antigen-presenting cells to mount sustained anti-tumor immune responses have been clinically evaluated. Further, senolytics that selectively deplete senescent cells are being tested in a number of clinical trials. Here we discuss the potential use of such drugs to improve anti-aging therapy.
Collapse
Affiliation(s)
- Christoph Hieber
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
30
|
Li D, Liu Q, Yang M, Xu H, Zhu M, Zhang Y, Xu J, Tian C, Yao J, Wang L, Liang Y. Nanomaterials for mRNA-based therapeutics: Challenges and opportunities. Bioeng Transl Med 2023; 8:e10492. [PMID: 37206219 PMCID: PMC10189457 DOI: 10.1002/btm2.10492] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/31/2023] Open
Abstract
Messenger RNA (mRNA) holds great potential in developing immunotherapy, protein replacement, and genome editing. In general, mRNA does not have the risk of being incorporated into the host genome and does not need to enter the nucleus for transfection, and it can be expressed even in nondividing cells. Therefore, mRNA-based therapeutics provide a promising strategy for clinical treatment. However, the efficient and safe delivery of mRNA remains a crucial constraint for the clinical application of mRNA therapeutics. Although the stability and tolerability of mRNA can be enhanced by directly retouching the mRNA structure, there is still an urgent need to improve the delivery of mRNA. Recently, significant progress has been made in nanobiotechnology, providing tools for developing mRNA nanocarriers. Nano-drug delivery system is directly used for loading, protecting, and releasing mRNA in the biological microenvironment and can be used to stimulate the translation of mRNA to develop effective intervention strategies. In the present review, we summarized the concept of emerging nanomaterials for mRNA delivery and the latest progress in enhancing the function of mRNA, primarily focusing on the role of exosomes in mRNA delivery. Moreover, we outlined its clinical applications so far. Finally, the key obstacles of mRNA nanocarriers are emphasized, and promising strategies to overcome these obstacles are proposed. Collectively, nano-design materials exert functions for specific mRNA applications, provide new perception for next-generation nanomaterials, and thus revolution of mRNA technology.
Collapse
Affiliation(s)
- De‐feng Li
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Qi‐song Liu
- National Clinical Research Center for Infectious DiseasesShenzhen Third People's Hospital, Southern University of Science and TechnologyShenzhenChina
| | - Mei‐feng Yang
- Department of HematologyYantian District People's HospitalShenzhenGuangdongChina
| | - Hao‐ming Xu
- Department of Gastroenterology and HepatologyGuangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Min‐zheng Zhu
- Department of Gastroenterology and Hepatologythe Second Affiliated Hospital, School of Medicine, South China University of TechnologyGuangzhouGuangdongChina
| | - Yuan Zhang
- Department of Medical AdministrationHuizhou Institute of Occupational Diseases Control and PreventionHuizhouGuangdongChina
| | - Jing Xu
- Department of Gastroenterology and HepatologyGuangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of TechnologyGuangzhouChina
| | - Cheng‐mei Tian
- Department of EmergencyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Jun Yao
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Li‐sheng Wang
- Department of GastroenterologyShenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Yu‐jie Liang
- Department of Child and Adolescent PsychiatryShenzhen Kangning Hospital, Shenzhen Mental Health CenterShenzhenChina
- Affiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
| |
Collapse
|
31
|
Yang W, Cao J, Cheng H, Chen L, Yu M, Chen Y, Cui X. Nanoformulations targeting immune cells for cancer therapy: mRNA therapeutics. Bioact Mater 2023; 23:438-470. [PMCID: PMC9712057 DOI: 10.1016/j.bioactmat.2022.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
The approved worldwide use of two messenger RNA (mRNA) vaccines (BNT162b2 and mRNA-1273) in late 2020 has proven the remarkable success of mRNA therapeutics together with lipid nanoformulation technology in protecting people against coronaviruses during COVID-19 pandemic. This unprecedented and exciting dual strategy with nanoformulations and mRNA therapeutics in play is believed to be a promising paradigm in targeted cancer immunotherapy in future. Recent advances in nanoformulation technologies play a prominent role in adapting mRNA platform in cancer treatment. In this review, we introduce the biologic principles and advancements of mRNA technology, and chemistry fundamentals of intriguing mRNA delivery nanoformulations. We discuss the latest promising nano-mRNA therapeutics for enhanced cancer immunotherapy by modulation of targeted specific subtypes of immune cells, such as dendritic cells (DCs) at peripheral lymphoid organs for initiating mRNA cancer vaccine-mediated antigen specific immunotherapy, and DCs, natural killer (NK) cells, cytotoxic T cells, or multiple immunosuppressive immune cells at tumor microenvironment (TME) for reversing immune evasion. We highlight the clinical progress of advanced nano-mRNA therapeutics in targeted cancer therapy and provide our perspectives on future directions of this transformative integrated technology toward clinical implementation.
Collapse
Affiliation(s)
- Wei Yang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, PR China
| | - Jianwei Cao
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, PR China
| | - Hui Cheng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China,Corresponding author
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China,Corresponding author
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, PR China,Corresponding author
| |
Collapse
|
32
|
Hu Y, Zhang W, Chu X, Wang A, He Z, Si CL, Hu W. Dendritic cell-targeting polymer nanoparticle-based immunotherapy for cancer: A review. Int J Pharm 2023; 635:122703. [PMID: 36758880 DOI: 10.1016/j.ijpharm.2023.122703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Cancer immunity is dependent on dynamic interactions between T cells and dendritic cells (DCs). Polymer-based nanoparticles target DC receptors to improve anticancer immune responses. In this paper, DC surface receptors and their specific coupling natural ligands and antibodies are reviewed and compared. Moreover, reaction mechanisms are described, and the synergistic effects of immune adjuvants are demonstrated. Also, extracellular-targeting antigen-delivery strategies and intracellular stimulus responses are reviewed to promote the rational design of polymer delivery systems.
Collapse
Affiliation(s)
- Yeye Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Tianjin Key Laboratory of Pulp & Paper, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wei Zhang
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Xiaozhong Chu
- School of Chemistry & Chemical Engineering, Huaiyin Normal University, Huaian 223300, China
| | - Aoran Wang
- School of Chemistry & Chemical Engineering, Huaiyin Normal University, Huaian 223300, China
| | - Ziliang He
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Chuan-Ling Si
- Tianjin Key Laboratory of Pulp & Paper, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Affiliated Hospital of Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
33
|
Li J, Lu W, Yang Y, Xiang R, Ling Y, Yu C, Zhou Y. Hybrid Nanomaterials for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204932. [PMID: 36567305 PMCID: PMC9951325 DOI: 10.1002/advs.202204932] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/30/2022] [Indexed: 06/17/2023]
Abstract
Nano-immunotherapy has been recognized as a highly promising strategy for cancer treatment in recent decades, which combines nanotechnology and immunotherapy to combat against tumors. Hybrid nanomaterials consisting of at least two constituents with distinct compositions and properties, usually organic and inorganic, have been engineered with integrated functions and enormous potential in boosting cancer immunotherapy. This review provides a summary of hybrid nanomaterials reported for cancer immunotherapy, including nanoscale metal-organic frameworks, metal-phenolic networks, mesoporous organosilica nanoparticles, metallofullerene nanomaterials, polymer-lipid, and biomacromolecule-based hybrid nanomaterials. The combination of immunotherapy with chemotherapy, chemodynamic therapy, radiotherapy, radiodynamic therapy, photothermal therapy, photodynamic therapy, and sonodynamic therapy based on hybrid nanomaterials is also discussed. Finally, the current challenges and the prospects for designing hybrid nanomaterials and their application in cancer immunotherapy are outlined.
Collapse
Affiliation(s)
- Jianing Li
- Shanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsDepartment of ChemistryFudan UniversityShanghai200433China
| | - Wanyue Lu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsDepartment of ChemistryFudan UniversityShanghai200433China
| | - Yannan Yang
- Institute of OptoelectronicsFudan UniversityShanghai200433China
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt LuciaBrisbane4072Australia
| | - Ruiqing Xiang
- Shanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsDepartment of ChemistryFudan UniversityShanghai200433China
| | - Yun Ling
- Shanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsDepartment of ChemistryFudan UniversityShanghai200433China
| | - Chengzhong Yu
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt LuciaBrisbane4072Australia
| | - Yaming Zhou
- Shanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsDepartment of ChemistryFudan UniversityShanghai200433China
| |
Collapse
|
34
|
mRNA-From COVID-19 Treatment to Cancer Immunotherapy. Biomedicines 2023; 11:biomedicines11020308. [PMID: 36830845 PMCID: PMC9953480 DOI: 10.3390/biomedicines11020308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
This review provides an overview covering mRNA from its use in the COVID-19 pandemic to cancer immunotherapy, starting from the selection of appropriate antigens, tumor-associated and tumor-specific antigens, neoantigens, the basics of optimizing the mRNA molecule in terms of stability, efficacy, and tolerability, choosing the best formulation and the optimal route of administration, to summarizing current clinical trials of mRNA vaccines in tumor therapy.
Collapse
|
35
|
Huang P, Jiang L, Pan H, Ding L, Zhou B, Zhao M, Zou J, Li B, Qi M, Deng H, Zhou Y, Chen X. An Integrated Polymeric mRNA Vaccine without Inflammation Side Effects for Cellular Immunity Mediated Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207471. [PMID: 36326183 DOI: 10.1002/adma.202207471] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/22/2022] [Indexed: 06/16/2023]
Abstract
Among the few available mRNA delivery vehicles, lipid nanoparticles (LNPs) are the most clinically advanced but they require cumbersome four components and suffer from inflammation-related side effects that should be minimized for safety. Yet, a certain level of proinflammatory responses and innate immune activation are required to evoke T-cell immunity for mRNA cancer vaccination. To address these issues and develop potent yet low-inflammatory mRNA cancer vaccine vectors, a series of alternating copolymers "PHTA" featured with ortho-hydroxy tertiary amine (HTA) repeating units for mRNA delivery is synthesized, which can play triple roles of condensing mRNA, enhancing the polymeric nanoparticle (PNP) stability, and prolonging circulation time. Unlike LNPs exhibiting high levels of inflammation, the PHTA-based PNPs show negligible inflammatory side effects in vivo. Importantly, the top candidate PHTA-C18 enables successful mRNA cancer vaccine delivery in vivo and leads to a robust CD8+ T cell mediated antitumor cellular immunity. Such PHTA-based integrated PNP provides a potential approach for establishing mRNA cancer vaccines with good inflammatory safety profiles.
Collapse
Affiliation(s)
- Pei Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Lingsheng Jiang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hui Pan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lingwen Ding
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
| | - Bo Zhou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Mengyao Zhao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Benhao Li
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Meiwei Qi
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hongzhang Deng
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Yongfeng Zhou
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
36
|
Fan N, Chen K, Zhu R, Zhang Z, Huang H, Qin S, Zheng Q, He Z, He X, Xiao W, Zhang Y, Gu Y, Zhao C, Liu Y, Jiang X, Li S, Wei Y, Song X. Manganese-coordinated mRNA vaccines with enhanced mRNA expression and immunogenicity induce robust immune responses against SARS-CoV-2 variants. SCIENCE ADVANCES 2022; 8:eabq3500. [PMID: 36563159 PMCID: PMC9788765 DOI: 10.1126/sciadv.abq3500] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
It is urgent to develop more effective mRNA vaccines against the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants owing to the immune escape. Here, we constructed a novel mRNA delivery system [IC8/Mn lipid nanoparticles (IC8/Mn LNPs)]with high immunogenicity, via introducing a stimulator of interferon genes (STING) agonist [manganese (Mn)] based on a newly synthesized ionizable lipid (IC8). It was found that Mn can not only promote maturation of antigen-presenting cells via activating STING pathway but also improve mRNA expression by facilitating lysosomal escape for the first time. Subsequently, IC8/Mn LNPs loaded with mRNA encoding the Spike protein of SARS-CoV-2 Delta or Omicron variant (IC8/Mn@D or IC8/Mn@O) were prepared. Both mRNA vaccines induced substantial specific immunoglobulin G responses against Delta or Omicron. IC8/Mn@D displayed strong pseudovirus neutralization ability, T helper 1-biased immune responses, and good safety. It can be concluded that IC8/Mn LNPs have great potential for developing Mn-coordinated mRNA vaccines with robust immunogenicity and good safety.
Collapse
Affiliation(s)
- Na Fan
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kepan Chen
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Zhu
- WestChina-Frontier PharmaTech Co. Ltd., Chengdu, Sichuan, China
| | - Zhongwei Zhang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai Huang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shugang Qin
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zheng
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongshan He
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xi He
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen Xiao
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yupei Zhang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongjun Gu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changchun Zhao
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongmei Liu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Jiang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuaicheng Li
- Department of Computer Science, City University of Hong Kong, Tat Chee Ave., Kowloon Tong, Hong Kong, China
| | - Yuquan Wei
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Corresponding author.
| |
Collapse
|
37
|
Stoitzner P, Romani N, Rademacher C, Probst HC, Mahnke K. Antigen targeting to dendritic cells: Still a place in future immunotherapy? Eur J Immunol 2022; 52:1909-1924. [PMID: 35598160 PMCID: PMC10084009 DOI: 10.1002/eji.202149515] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/01/2022] [Accepted: 05/20/2022] [Indexed: 12/16/2022]
Abstract
The hallmark of DCs is their potent and outstanding capacity to activate naive resting T cells. As such, DCs are the sentinels of the immune system and instrumental for the induction of immune responses. This is one of the reasons, why DCs became the focus of immunotherapeutical strategies to fight infections, cancer, and autoimmunity. Besides the exploration of adoptive DC-therapy for which DCs are generated from monocytes or purified in large numbers from the blood, alternative approaches were developed such as antigen targeting of DCs. The idea behind this strategy is that DCs resident in patients' lymphoid organs or peripheral tissues can be directly loaded with antigens in situ. The proof of principle came from mouse models; subsequent translational studies confirmed the potential of this therapy. The first clinical trials demonstrated feasibility and the induction of T-cell immunity in patients. This review will cover: (i) the historical aspects of antigen targeting, (ii) briefly summarize the biology of DCs and the immunological functions upon which this concept rests, (iii) give an overview on attempts to target DC receptors with antibodies or (glycosylated) ligands, and finally, (iv) discuss the translation of antigen targeting into clinical therapy.
Collapse
Affiliation(s)
- Patrizia Stoitzner
- Department of Dermatology, Venereology, and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nikolaus Romani
- Department of Dermatology, Venereology, and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Rademacher
- Department of Microbiology, Immunology and Genetics, University of Vienna, Vienna, Austria.,Institute of Immunology, University Medical Center Mainz, Mainz, Germany
| | - Hans Christian Probst
- Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany.,Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Karsten Mahnke
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
38
|
Valenzuela-Fernández A, Cabrera-Rodriguez R, Ciuffreda L, Perez-Yanes S, Estevez-Herrera J, González-Montelongo R, Alcoba-Florez J, Trujillo-González R, García-Martínez de Artola D, Gil-Campesino H, Díez-Gil O, Lorenzo-Salazar JM, Flores C, Garcia-Luis J. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Front Bioeng Biotechnol 2022; 10:1052436. [PMID: 36507266 PMCID: PMC9732709 DOI: 10.3389/fbioe.2022.1052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the associated coronavirus disease 2019 (COVID-19), which severely affect the respiratory system and several organs and tissues, and may lead to death, have shown how science can respond when challenged by a global emergency, offering as a response a myriad of rapid technological developments. Development of vaccines at lightning speed is one of them. SARS-CoV-2 outbreaks have stressed healthcare systems, questioning patients care by using standard non-adapted therapies and diagnostic tools. In this scenario, nanotechnology has offered new tools, techniques and opportunities for prevention, for rapid, accurate and sensitive diagnosis and treatment of COVID-19. In this review, we focus on the nanotechnological applications and nano-based materials (i.e., personal protective equipment) to combat SARS-CoV-2 transmission, infection, organ damage and for the development of new tools for virosurveillance, diagnose and immune protection by mRNA and other nano-based vaccines. All the nano-based developed tools have allowed a historical, unprecedented, real time epidemiological surveillance and diagnosis of SARS-CoV-2 infection, at community and international levels. The nano-based technology has help to predict and detect how this Sarbecovirus is mutating and the severity of the associated COVID-19 disease, thereby assisting the administration and public health services to make decisions and measures for preparedness against the emerging variants of SARS-CoV-2 and severe or lethal COVID-19.
Collapse
Affiliation(s)
- Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Romina Cabrera-Rodriguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Laura Ciuffreda
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Perez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Judith Estevez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | - Julia Alcoba-Florez
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departamento de Análisis Matemático, Facultad de Ciencias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Helena Gil-Campesino
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Oscar Díez-Gil
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Jonay Garcia-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
39
|
Markov OV, Sen’kova AV, Mohamed IS, Shmendel EV, Maslov MA, Oshchepkova AL, Brenner EV, Mironova NL, Zenkova MA. Dendritic Cell-Derived Artificial Microvesicles Inhibit RLS 40 Lymphosarcoma Growth in Mice via Stimulation of Th1/Th17 Immune Response. Pharmaceutics 2022; 14:pharmaceutics14112542. [PMID: 36432733 PMCID: PMC9696603 DOI: 10.3390/pharmaceutics14112542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Cell-free antitumor vaccines represent a promising approach to immunotherapy of cancer. Here, we compare the antitumor potential of cell-free vaccines based on microvesicles derived from dendritic cells (DCs) with DC- and cationic-liposome-based vaccines using a murine model of drug-resistant lymphosarcoma RLS40 in vivo. The vaccines were the following: microvesicle vaccines—cytochalasin B-induced membrane vesicles (CIMVs) obtained from DCs loaded with total tumor RNA using cholesterol/spermine-containing cationic liposomes L or mannosylated liposomes ML; DC vaccines—murine DCs loaded with total tumor-derived RNA using the same liposomes; and liposomal vaccines—lipoplexes of total tumor-derived RNA with liposomes L or ML. Being non-hepatotoxic, CIMV- and DC-based vaccines administered subcutaneously exhibited comparable potential to stimulate highly efficient antitumor CTLs in vivo, whereas liposomal vaccines were 25% weaker CTL inducers. Nevertheless, the antitumor efficiencies of the different types of the vaccines were similar: sizes of tumor nodes and the number of liver metastases were significantly decreased, regardless of the vaccine type. Notably, the booster vaccination did not improve the overall antitumor efficacy of the vaccines under the study. CIMV- and DC- based vaccines more efficiently than liposome-based ones decreased mitotic activity of tumor cells and induced their apoptosis, stimulated accumulation of neutrophil inflammatory infiltration in tumor tissue, and had a more pronounced immunomodulatory activity toward the spleen and thymus. Administration of CIMV-, DC-, and liposome-based vaccines resulted in activation of Th1/Th17 cells as well as the induction of positive immune checkpoint 4-1BBL and downregulation of suppressive immune checkpoints in a raw PD-1 >>> TIGIT > CTLA4 > TIM3. We demonstrated that cell-free CIMV-based vaccines exhibited superior antitumor and antimetastatic activity in a tumor model in vivo. The obtained results can be considered as the basis for developing novel strategies for oncoimmunotherapy.
Collapse
Affiliation(s)
- Oleg V. Markov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-(383)-363-51-61
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia
| | - Islam S. Mohamed
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia
| | - Elena V. Shmendel
- M.V. Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo Ave. 86, 119571 Moscow, Russia
| | - Mikhail A. Maslov
- M.V. Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo Ave. 86, 119571 Moscow, Russia
| | - Anastasiya L. Oshchepkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia
| | - Evgeniy V. Brenner
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia
| | - Nadezhda L. Mironova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, 630090 Novosibirsk, Russia
| |
Collapse
|
40
|
Diep YN, Kim TJ, Cho H, Lee LP. Nanomedicine for advanced cancer immunotherapy. J Control Release 2022; 351:1017-1037. [DOI: 10.1016/j.jconrel.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/09/2022]
|
41
|
Li Y, Fang H, Zhang T, Wang Y, Qi T, Li B, Jiao H. Lipid-mRNA nanoparticles landscape for cancer therapy. Front Bioeng Biotechnol 2022; 10:1053197. [PMID: 36394007 PMCID: PMC9659646 DOI: 10.3389/fbioe.2022.1053197] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 10/18/2022] [Indexed: 09/19/2023] Open
Abstract
Intracellular delivery of message RNA (mRNA) technique has ushered in a hopeful era with the successive authorization of two mRNA vaccines for the Coronavirus disease-19 (COVID-19) pandemic. A wide range of clinical studies are proceeding and will be initiated in the foreseeable future to treat and prevent cancers. However, efficient and non-toxic delivery of therapeutic mRNAs maintains the key limited step for their widespread applications in human beings. mRNA delivery systems are in urgent demand to resolve this difficulty. Recently lipid nanoparticles (LNPs) vehicles have prospered as powerful mRNA delivery tools, enabling their potential applications in malignant tumors via cancer immunotherapy and CRISPR/Cas9-based gene editing technique. This review discusses formulation components of mRNA-LNPs, summarizes the latest findings of mRNA cancer therapy, highlights challenges, and offers directions for more effective nanotherapeutics for cancer patients.
Collapse
Affiliation(s)
- Yin Li
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Hengtong Fang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Tao Zhang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Yu Wang
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Tingting Qi
- College of Animal Science, Jilin University, Changchun, Jilin, China
| | - Bai Li
- Department of Colorectal and Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huping Jiao
- College of Animal Science, Jilin University, Changchun, Jilin, China
| |
Collapse
|
42
|
Lopes C, Cristóvão J, Silvério V, Lino PR, Fonte P. Microfluidic production of mRNA-loaded lipid nanoparticles for vaccine applications. Expert Opin Drug Deliv 2022; 19:1381-1395. [PMID: 36223174 DOI: 10.1080/17425247.2022.2135502] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION During past years, lipid nanoparticles (LNPs) have emerged as promising carriers for RNA delivery, with several clinical trials focusing on both infectious diseases and cancer. More recently, the success of messenger RNA (mRNA) vaccines for the treatment of severe diseases such as acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is partially justified by the development of LNPs encapsulating mRNA for efficient cytosolic delivery. AREAS COVERED This review examines the production and formulation of LNPs by using microfluidic devices, the status of mRNA-loaded LNPs therapeutics and explores spray drying process, as a promising dehydration process to enhance LNP stability and provide alternative administration routes. EXPERT OPINION Microfluidic techniques for preparation of LNPs based on organic solvent injection method promotes the generation of stable, uniform, and monodispersed nanoparticles enabling higher encapsulation efficiency. In particular, the application of microfluidics for the fabrication of mRNA-loaded LNPs is based on rapid mixing of small volumes of ethanol solution containing lipids and aqueous solution containing mRNA. Control of operating parameters and formulation has enabled the optimization of nanoparticle physicochemical characteristics and encapsulation efficiency.
Collapse
Affiliation(s)
- Carolina Lopes
- iBB - Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Hovione Farmaciência S.A., R&D Analytical Development, Lumiar Campus, Building R,1649-038 Lisbon, Portugal.,Hovione Farmaciência S.A., R&D Inhalation and Advance Drug Delivery, Lumiar Campus, Building R, 1649-038 Lisbon, Portugal
| | - Joana Cristóvão
- Hovione Farmaciência S.A., R&D Inhalation and Advance Drug Delivery, Lumiar Campus, Building R, 1649-038 Lisbon, Portugal
| | - Vânia Silvério
- Institute of Systems and Computer Engineering for Microsystems and Nanotechnologies, INESC MN, 1000-029 Lisbon, Portugal.,Department of Physics, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal
| | - Paulo Roque Lino
- Hovione Farmaciência S.A., R&D Inhalation and Advance Drug Delivery, Lumiar Campus, Building R, 1649-038 Lisbon, Portugal
| | - Pedro Fonte
- iBB - Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.,Center of Marine Sciences (CCMAR), University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal.,Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
| |
Collapse
|
43
|
RNA modifications: importance in immune cell biology and related diseases. Signal Transduct Target Ther 2022; 7:334. [PMID: 36138023 PMCID: PMC9499983 DOI: 10.1038/s41392-022-01175-9] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
RNA modifications have become hot topics recently. By influencing RNA processes, including generation, transportation, function, and metabolization, they act as critical regulators of cell biology. The immune cell abnormality in human diseases is also a research focus and progressing rapidly these years. Studies have demonstrated that RNA modifications participate in the multiple biological processes of immune cells, including development, differentiation, activation, migration, and polarization, thereby modulating the immune responses and are involved in some immune related diseases. In this review, we present existing knowledge of the biological functions and underlying mechanisms of RNA modifications, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), N7-methylguanosine (m7G), N4-acetylcytosine (ac4C), pseudouridine (Ψ), uridylation, and adenosine-to-inosine (A-to-I) RNA editing, and summarize their critical roles in immune cell biology. Via regulating the biological processes of immune cells, RNA modifications can participate in the pathogenesis of immune related diseases, such as cancers, infection, inflammatory and autoimmune diseases. We further highlight the challenges and future directions based on the existing knowledge. All in all, this review will provide helpful knowledge as well as novel ideas for the researchers in this area.
Collapse
|
44
|
Huang T, Peng L, Han Y, Wang D, He X, Wang J, Ou C. Lipid nanoparticle-based mRNA vaccines in cancers: Current advances and future prospects. Front Immunol 2022; 13:922301. [PMID: 36090974 PMCID: PMC9458914 DOI: 10.3389/fimmu.2022.922301] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/08/2022] [Indexed: 12/24/2022] Open
Abstract
Messenger RNA (mRNA) vaccines constitute an emerging therapeutic method with the advantages of high safety and efficiency as well as easy synthesis; thus, they have been widely used in various human diseases, especially in malignant cancers. However, the mRNA vaccine technology has some limitations, such as instability and low transitive efficiency in vivo, which greatly restrict its application. The development of nanotechnology in the biomedical field offers new strategies and prospects for the early diagnosis and treatment of human cancers. Recent studies have demonstrated that Lipid nanoparticle (LNP)-based mRNA vaccines can address the poor preservation and targeted inaccuracy of mRNA vaccines. As an emerging cancer therapy, mRNA vaccines potentially have broad future applications. Unlike other treatments, cancer mRNA vaccines provide specific, safe, and tolerable treatments. Preclinical studies have used personalized vaccines to demonstrate the anti-tumor effect of mRNA vaccines in the treatment of various solid tumors, including colorectal and lung cancer, using these in a new era of therapeutic cancer vaccines. In this review, we have summarized the latest applications and progress of LNP-based mRNA vaccines in cancers, and discussed the prospects and limitations of these fields, thereby providing novel strategies for the targeted therapy of cancers.
Collapse
Affiliation(s)
- Tao Huang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Lushan Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yingying Han
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Dan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, China
| | - Junpu Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
Shi Y, Lu Y, You J. Antigen transfer and its effect on vaccine-induced immune amplification and tolerance. Am J Cancer Res 2022; 12:5888-5913. [PMID: 35966588 PMCID: PMC9373810 DOI: 10.7150/thno.75904] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/15/2022] [Indexed: 12/13/2022] Open
Abstract
Antigen transfer refers to the process of intercellular information exchange, where antigenic components including nucleic acids, antigen proteins/peptides and peptide-major histocompatibility complexes (p-MHCs) are transmitted from donor cells to recipient cells at the thymus, secondary lymphoid organs (SLOs), intestine, allergic sites, allografts, pathological lesions and vaccine injection sites via trogocytosis, gap junctions, tunnel nanotubes (TNTs), or extracellular vesicles (EVs). In the context of vaccine inoculation, antigen transfer is manipulated by the vaccine type and administration route, which consequently influences, even alters the immunological outcome, i.e., immune amplification and tolerance. Mainly focused on dendritic cells (DCs)-based antigen receptors, this review systematically introduces the biological process, molecular basis and clinical manifestation of antigen transfer.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
46
|
Bevers S, Kooijmans SAA, Van de Velde E, Evers MJW, Seghers S, Gitz-Francois JJJM, van Kronenburg NCH, Fens MHAM, Mastrobattista E, Hassler L, Sork H, Lehto T, Ahmed KE, El Andaloussi S, Fiedler K, Breckpot K, Maes M, Van Hoorick D, Bastogne T, Schiffelers RM, De Koker S. mRNA-LNP vaccines tuned for systemic immunization induce strong antitumor immunity by engaging splenic immune cells. Mol Ther 2022; 30:3078-3094. [PMID: 35821637 PMCID: PMC9273295 DOI: 10.1016/j.ymthe.2022.07.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/23/2022] [Accepted: 07/09/2022] [Indexed: 12/19/2022] Open
Abstract
mRNA vaccines have recently proven to be highly effective against SARS-CoV-2. Key to their success is the lipid-based nanoparticle (LNP), which enables efficient mRNA expression and endows the vaccine with adjuvant properties that drive potent antibody responses. Effective cancer vaccines require long-lived, qualitative CD8 T cell responses instead of antibody responses. Systemic vaccination appears to be the most effective route, but necessitates adaptation of LNP composition to deliver mRNA to antigen presenting cells. Using a design-of-experiments methodology, we tailored mRNA-LNP compositions to achieve high magnitude tumor-specific CD8 T cell responses within a single round of optimization. Optimized LNP compositions resulted in enhanced mRNA uptake by multiple splenic immune cell populations. Type I interferon and phagocytes were found essential for the T cell response. Surprisingly, we also discovered a yet unidentified role of B cells in stimulating the vaccine-elicited CD8 T cell response. Optimized LNPs displayed a similar, spleen-centered biodistribution profile in non-human primates and did not trigger histopathological changes in liver and spleen, warranting their further assessment in clinical studies. Taken together, our study clarifies the relationship between nanoparticle composition and their T cell stimulatory capacity and provides novel insights into the underlying mechanisms of effective mRNA-LNP based antitumor immunotherapy.
Collapse
Affiliation(s)
- Sanne Bevers
- eTheRNA Immunotherapies, 2845 Niel, Belgium; Laboratory for Molecular and Cellular Therapy (LMCT), Free University of Brussels, 1090 Jette, Belgium
| | - Sander A A Kooijmans
- CDL Research, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | | | - Martijn J W Evers
- CDL Research, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | | | | | - Nicky C H van Kronenburg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Marcel H A M Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, the Netherlands
| | | | - Helena Sork
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Taavi Lehto
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia; Department of Laboratory Medicine, Karolinksa Institutet, 141 52 Huddinge, Sweden
| | - Kariem E Ahmed
- Department of Laboratory Medicine, Karolinksa Institutet, 141 52 Huddinge, Sweden
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinksa Institutet, 141 52 Huddinge, Sweden
| | | | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Free University of Brussels, 1090 Jette, Belgium
| | | | | | - Thierry Bastogne
- CYBERnano, 54000 Nancy, France; CRAN, Université de Lorraine, CNRS, INRIA BIGS, 54506 Vandœuvre-lès-Nancy, France
| | | | | |
Collapse
|
47
|
Zhang M, Hussain A, Yang H, Zhang J, Liang XJ, Huang Y. mRNA-based modalities for infectious disease management. NANO RESEARCH 2022; 16:672-691. [PMID: 35818566 PMCID: PMC9258466 DOI: 10.1007/s12274-022-4627-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
The novel coronavirus disease 2019 (COVID-19) is still rampant all over the world, causing incalculable losses to the world. Major pharmaceutical organizations around the globe are focusing on vaccine research and drug development to prevent further damage caused by the pandemic. The messenger RNA (mRNA) technology has got ample of attention after the success of the two very effective mRNA vaccines during the recent pandemic of COVID-19. mRNA vaccine has been promoted to the core stage of pharmaceutical industry, and the rapid development of mRNA technology has exceeded expectations. Beyond COVID-19, the mRNA vaccine has been tested for various infectious diseases and undergoing clinical trials. Due to the ability of constant mutation, the viral infections demand abrupt responses and immediate production, and therefore mRNA-based technology offers best answers to sudden outbreaks. The need for mRNA-based vaccine became more obvious due to the recent emergence of new Omicron variant. In this review, we summarized the unique properties of mRNA-based vaccines for infectious diseases, delivery technologies, discussed current challenges, and highlighted the prospects of this promising technology in the future. We also discussed various clinical studies as well preclinical studies conducted on mRNA therapeutics for diverse infectious diseases.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry and Environmental Science, Hebei University, Baoding, 071002 China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081 China
- School of Materials and the Environment, Beijing Institute of Technology, Zhuhai, 519085 China
| |
Collapse
|
48
|
Nakamura T, Sato Y, Yamada Y, Abd Elwakil MM, Kimura S, Younis MA, Harashima H. Extrahepatic targeting of lipid nanoparticles in vivo with intracellular targeting for future nanomedicines. Adv Drug Deliv Rev 2022; 188:114417. [PMID: 35787389 DOI: 10.1016/j.addr.2022.114417] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/02/2022] [Accepted: 06/28/2022] [Indexed: 12/15/2022]
Abstract
A new era of nanomedicines that involve nucleic acids/gene therapy has been opened after two decades in 21st century and new types of more efficient drug delivery systems (DDS) are highly expected and will include extrahepatic delivery. In this review, we summarize the possibility and expectations for the extrahepatic delivery of small interfering RNA/messenger RNA/plasmid DNA/genome editing to the spleen, lung, tumor, lymph nodes as well as the liver based on our studies as well as reported information. Passive targeting and active targeting are discussed in in vivo delivery and the importance of controlled intracellular trafficking for successful therapeutic results are also discussed. In addition, mitochondrial delivery as a novel strategy for nucleic acids/gene therapy is introduced to expand the therapeutic dimension of nucleic acids/gene therapy in the liver as well as the heart, kidney and brain.
Collapse
Affiliation(s)
- Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Mahmoud M Abd Elwakil
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Seigo Kimura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Mahmoud A Younis
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
49
|
Mokuda S, Watanabe H, Kohno H, Ishitoku M, Araki K, Hirata S, Sugiyama E. N1-methylpseudouridine-incorporated mRNA enhances exogenous protein expression and suppresses immunogenicity in primary human fibroblast-like synoviocytes. Cytotechnology 2022; 74:503-514. [PMID: 35791402 PMCID: PMC9245880 DOI: 10.1007/s10616-022-00540-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/03/2022] [Indexed: 11/25/2022] Open
Abstract
Studies conducted using murine arthritis models have indicated that the use of in vitro-transcribed messenger RNA (IVT mRNA) is an effective therapeutic approach for joint diseases. However, the use of IVT mRNA in human synovial cells has not been widely studied. Recently, the outbreak of the novel coronavirus disease has accelerated the development of innovative mRNA vaccines, such as those containing a modified nucleic acid, N1-methylpseudouridine-5′-triphosphate (m1ψ). IVT mRNA is an attractive tool for biological experiments and drug discovery. To verify the protein expression from IVT mRNA in vitro, primary cultured fibroblast-like synoviocytes (FLS) and MH7A human synovial fibroblast cells were transfected with enhanced green fluorescent protein (EGFP) mRNA with or without m1ψ incorporation. EGFP was detected using western blotting and fluorescence microscopy. A multiplex assay was performed to comprehensively understand IVT mRNA-induced immunogenicity. Gene expression levels were measured using reverse transcription polymerase chain reaction. In both MH7A cells and FLS, cells transfected with EGFP mRNA containing m1ψ generated higher levels of EGFP than those transfected with unmodified EGFP or control mRNAs. The multiplex assay of the FLS culture supernatant and reverse transcription polymerase chain reaction for FLS revealed that both concentration and expression of IL-6, TNF-α, and CXCL10 were upregulated by unmodified EGFP mRNA, whereas they were suppressed by EGFP mRNA with m1ψ. Overall, m1ψ incorporation enhanced protein expression and decreased the expression of cytokines. These findings may contribute to arthritis research.
Collapse
Affiliation(s)
- Sho Mokuda
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Hirofumi Watanabe
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Hiroki Kohno
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Michinori Ishitoku
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kei Araki
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shintaro Hirata
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Eiji Sugiyama
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
50
|
D’haese S, Laeremans T, den Roover S, Allard SD, Vanham G, Aerts JL. Efficient Induction of Antigen-Specific CD8+ T-Cell Responses by Cationic Peptide-Based mRNA Nanoparticles. Pharmaceutics 2022; 14:pharmaceutics14071387. [PMID: 35890284 PMCID: PMC9321026 DOI: 10.3390/pharmaceutics14071387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
A major determinant for the success of mRNA-based vaccines is the composition of the nanoparticles (NPs) used for formulation and delivery. Cationic peptides represent interesting candidate carriers for mRNA, since they have been shown to efficiently deliver nucleic acids to eukaryotic cells. mRNA NPs based on arginine-rich peptides have previously been demonstrated to induce potent antigen-specific CD8+ T-cell responses. We therefore compared the histidine-rich amphipathic peptide LAH4-L1 (KKALLAHALHLLALLALHLAHALKKA) to the fully substituted arginine variant (LAH4-L1R) for their capacity to formulate mRNA and transfect dendritic cells (DCs). Although both peptides encapsulated mRNA to the same extent, and showed excellent uptake in DCs, the gene expression level was significantly higher for LAH4-L1. The LAH4-L1–mRNA NPs also resulted in enhanced antigen presentation in the context of MHC I compared to LAH4-L1R in primary murine CD103+ DCs. Both peptides induced DC maturation and inflammasome activation. Subsequent ex vivo stimulation of OT-I splenocytes with transfected CD103+ DCs resulted in a high proportion of polyfunctional CD8+ T cells for both peptides. In addition, in vivo immunization with LAH4-L1 or LAH4-L1R–mRNA NPs resulted in proliferation of antigen-specific T cells. In conclusion, although LAH4-L1 outperformed LAH4-L1R in terms of transfection efficiency, the immune stimulation ex vivo and in vivo was equally efficient.
Collapse
Affiliation(s)
- Sigrid D’haese
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
| | - Thessa Laeremans
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
| | - Sabine den Roover
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
| | - Sabine D. Allard
- Department of Internal Medicine (IRG), Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium;
| | - Guido Vanham
- Department of Virology, Institute of Tropical Medicine, University of Antwerp, 2000 Antwerp, Belgium;
| | - Joeri L. Aerts
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (S.D.); (T.L.); (S.d.R.)
- Correspondence:
| |
Collapse
|