1
|
Amor NP, Guo K, Zhang S, Xia J, Yang Y, Lin A. Lipid Nanoparticle: Beyond Delivery Vehicle-Unveiling Its Immunological Adjuvant Potential. FASEB J 2025; 39:e70641. [PMID: 40372384 DOI: 10.1096/fj.202500622r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025]
Abstract
Lipid nanoparticles (LNPs) have extensively been used in drug delivery over the years, and the perspective of their significance has been well established. Latest findings have demonstrated the inherent adjuvant capacity of some specific lipid components, especially in stimulating immune compartments, which prompted the rational use of lipid-based delivery vehicles in drug R&D. In this concise review, we summarize current knowledge on the adjuvant properties of LNP, with a particular focus on the key components that mediate such effects. Specifically, we describe the vital role of ionizable lipids in triggering innate immune activation and inflammation and highlight the importance of these lipids in determining vaccine effectiveness or safety. Furthermore, the mechanisms by which LNP enhance the immune response are discussed in detail, shedding light on their potential applications in next-generation vaccine design and development. We also present compelling pre-clinical studies that serve as strong evidence for the adjuvant properties of LNP components in enhancing vaccine immunogenicity.
Collapse
Affiliation(s)
- Narh Philip Amor
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kun Guo
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shun Zhang
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Jun Xia
- Institute of Veterinary Medicine, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Yong Yang
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ang Lin
- Innovation Center for Nucleic Acid Medicine, Institute for Innovative Drug Development and Life Sciences, Wuxi, China Pharmaceutical University, Nanjing, China
- Institute of Translational Medicine, China Pharmaceutical University, Nanjing, China
- Center for Infectious Medicine and Vaccine Research, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
2
|
Berber E, Pantouli F, Hanley HB, Ross TM. COVID-19 Vaccination Enhances the Immunogenicity of Seasonal Influenza Vaccination in the Elderly. Vaccines (Basel) 2025; 13:531. [PMID: 40432140 PMCID: PMC12116172 DOI: 10.3390/vaccines13050531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The co-circulation of both influenza viruses and SARS-CoV-2 poses a significant health risk, especially for the elderly. While vaccination against both diseases remains an effective strategy to reduce the burden of symptomatic infections, the effect of administering COVID-19 mRNA and seasonal influenza vaccines (COV-Flu) on elicited antibody responses has not been explored. METHODS Participants between 18 and 90 years old were vaccinated with COVID-19 mRNA vaccines (n = 67), seasonal influenza vaccines (n = 130), or both (n = 201) within a three-month period between 2021 and 2024. Serum hemagglutination-inhibition (HAI) titers against influenza A (H1N1, H3N2) and B (Yamagata, Victoria) strains were measured from the COV-Flu participants or the participants vaccinated with influenza vaccines only (mono-Flu). SARS-CoV-2 neutralization assays were performed on sera collected from the COV-Flu participants and the participants receiving the mRNA vaccine only (mono-COVID-19). RESULTS The administration of influenza virus vaccines and COVID-19 mRNA vaccines within a three-month period significantly enhanced the post-vaccination HAI titers against both influenza A and B vaccine components, particularly in the elderly (65-90) participants. There were no significant differences in SARS-CoV-2 neutralization titers in COV-Flu participants compared to mono-COVID-19 participants. CONCLUSIONS Vaccination with both the COVID-19 mRNA and influenza vaccines enhances influenza-specific HAI titers without compromising the neutralization titers elicited by COVID-19 mRNA vaccination against SARS-CoV-2, especially in the elderly. These findings indicate the potential benefits of this approach, particularly for older adults, by boosting influenza virus vaccine-induced serum HAI activity while maintaining COVID-19 protective immunity.
Collapse
Affiliation(s)
- Engin Berber
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Fani Pantouli
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA;
| | - Hannah B. Hanley
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
| | - Ted M. Ross
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA;
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
3
|
O'Brien Laramy M, Foley DA, Pak RH, Lewis JA, McKinney E, Egan PM, Yerabolu R, Dane E, Dirat O, Saunders Gorka L, Martinelli JR, Moussa EM, Barthuet J. Chemistry, manufacturing and controls strategies for using novel excipients in lipid nanoparticles. NATURE NANOTECHNOLOGY 2025; 20:331-344. [PMID: 39821140 DOI: 10.1038/s41565-024-01833-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/30/2024] [Indexed: 01/19/2025]
Abstract
Lipid nanoparticles (LNPs) for nucleic acid delivery often use novel lipids as functional excipients to modulate the biodistribution, pharmacokinetics, pharmacodynamics and efficacy of the nucleic acid. Novel excipients used in pharmaceutical products are subject to heightened regulatory scrutiny and often require data packages comparable to an active pharmaceutical ingredient. Although these regulatory requirements may help to ensure patient safety they also create economic and procedural barriers that can disincentivize innovation and delay clinical investigation. Despite the unique structural and functional role of lipid excipients in LNPs, there is limited specific global regulatory guidance, which adds uncertainty and risk to the development of LNPs. In this Perspective we provide an industry view on the chemistry, manufacturing and controls challenges that pharmaceutical companies face in the use of novel lipid excipients at each stage of development, and propose consensus recommendations on how to streamline and clarify development and regulatory expectations.
Collapse
Affiliation(s)
- Matthew O'Brien Laramy
- Synthetic Molecule Pharmaceutical Sciences, Genentech Early Research and Development, Genentech, Inc., South San Francisco, CA, USA.
| | - David A Foley
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, USA.
| | - Roger H Pak
- Biotherapeutics Pharmaceutical Research and Development, Pfizer, Inc., Andover, MA, USA
| | - Jacob A Lewis
- Drug Product Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, USA
| | - Eric McKinney
- CMC Regulatory Affairs, Alnylam Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Patricia M Egan
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, USA
| | | | - Eric Dane
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, MA, USA
| | - Olivier Dirat
- Global Regulatory Sciences CMC Advisory Office, Pfizer, Inc., Sandwich, UK
| | | | | | - Ehab M Moussa
- Biologics Drug Product Development, AbbVie Inc., North Chicago, IL, USA
| | - Julie Barthuet
- Global Regulatory Affairs CMC, Sanofi, Marcy-l'Etoile, France
| |
Collapse
|
4
|
Ye L, Wang T, Wu S, Liu H, Liu F, Wang C, Hu M. Effects of immune response on different nano-adjuvants combined with H11 antigen of Haemonchus contortus. Int Immunopharmacol 2024; 143:113602. [PMID: 39536488 DOI: 10.1016/j.intimp.2024.113602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
The intestinal native protein H11 is one of the most immunodominant antigen of Haemonchus contortus. However, H11 combined with QuilA adjuvant shows only short-lived protection for animals. Nano-adjuvants have the huge potential to extend the immune response in human and animals. Here, we compared the immune responses induced by H11 combined with three nano-adjuvants including lipid nanoparticles (LNP), immunostimulating complex matrix (IMX) and AddaS03™. We measured the cytokine levels of goat PBMCs stimulated by H11 mixed with three nano-adjuvants and QuilA. Results showed that the transcriptions of IL-6, IL-8 and IFN-γ genes were significantly inhibited in all adjuvant group compared with PBS control. H11-IMX combination significantly up-regulated IL-2, IL-17 and TNF-α gene transcriptions. The H11-LNP group showed a significant increase in IL-17 and TNF-α transcriptions, while the H11-AddaS03™ group significantly increased IL-2 transcription. Additionally, mice immunized with these formulations were assessed for splenic lymphocyte proliferation. All H11-adjuvant combinations, except H11-LNP, significantly stimulated lymphocyte proliferation compared to the H11 alone and PBS control groups, with the H11-AddaS03™ combination showing the strongest effect. Analysis of serum antibody levels revealed that all immune groups induced high IgM levels, with H11-IMX inducing the highest IgG levels. Our results demonstrated that IMX or LNP combined with H11 mainly induced a Th17 cell immune response in goat PBMCs, while IMX or AddaS03™ combined with H11 could induce a strong humoral immune response in mice. This study elucidates the immune response profiles of different nano-adjuvant combined with H11 antigen, providing important insights for vaccine development against parasitic nematodes.
Collapse
Affiliation(s)
- Lisha Ye
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| | - Tianjiao Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| | - Simin Wu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| | - Hui Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| | - Feng Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| | - Chunqun Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| | - Min Hu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
5
|
Dai W, Xing M, Sun L, Lv L, Wang X, Wang Y, Pang X, Guo Y, Ren J, Zhou D. Lipid nanoparticles as adjuvant of norovirus VLP vaccine augment cellular and humoral immune responses in a TLR9- and type I IFN-dependent pathway. J Virol 2024; 98:e0169924. [PMID: 39494905 DOI: 10.1128/jvi.01699-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Norovirus (NoV) virus-like particles (VLPs) adjuvanted with aluminum hydroxide (Alum) are common vaccine candidates in clinical studies. Alum adjuvants usually inefficiently assist recombinant proteins to induce cellular immune responses. Thus, novel adjuvants are required to develop NoV vaccines that could induce both efficient humoral and robust cellular immune responses. Lipid nanoparticles (LNPs) are well-known mRNA delivery vehicles. Increasing evidence suggests that LNPs may have intrinsic adjuvant activity and can be used as adjuvants for recombinant protein vaccines; however, the underlying mechanism remains poorly understood. In this study, we compared the adjuvant effect of LNPs and Alum for a bivalent GI.1/GII.4 NoV VLP vaccine. Compared with Alum, LNP-adjuvanted vaccines induced earlier production of binding, blocking, and neutralizing antibodies and promoted a more balanced IgG2a/IgG1 ratio. It is crucial that LNP-adjuvanted vaccines induced stronger Th1-type cytokine-producing CD4+ T cell and CD8+ T cell responses than Alum. The adjuvant activity of LNPs depended on the ionizable lipid components. Mechanistically, LNPs activated innate immune responses in a type I IFN-dependent manner and were partially dependent on Toll-like receptor (TLR) 9, thus affecting the adaptive immune responses of the vaccine. This conclusion was supported by RNA-seq analysis and in vitro cell experiments and by the deeply blunted T cell responses in IFNαR1-/- mice immunized with LNP-adjuvanted vaccines. This study not only identified LNPs as a high quality adjuvant for NoV VLP vaccines, but also clarified the underlying mechanism of LNPs as a potent immunostimulatory component for improving protein subunit vaccines.IMPORTANCEWith the rapid development of mRNA vaccines, recurrent studies show that lipid nanoparticles (LNPs) have adjuvant activity. However, the mechanism of its adjuvant effect in protein vaccines remains unknown. In this study, we found that the LNP-adjuvanted norovirus bivalent virus-like particle vaccines led to durable antibody responses as well as Th1-type cytokine-producing CD4+ T cell and CD8+ T cell responses, which exceeded the efficiency of the conventional adjuvant aluminum hydroxide. Mechanistically, LNPs activated innate immune responses in a type I IFN-dependent manner and were partially dependent on Toll-like receptor 9, thus affecting the adaptive immune responses of the vaccine. This work unveils that LNPs as a potent immunostimulatory component may be ideal for generating CD8+ T cell and B cell responses for recombinant protein vaccines.
Collapse
MESH Headings
- Animals
- Nanoparticles/administration & dosage
- Mice
- Norovirus/immunology
- Immunity, Humoral
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Adjuvants, Immunologic/administration & dosage
- Interferon Type I/immunology
- Immunity, Cellular
- Antibodies, Viral/immunology
- Caliciviridae Infections/prevention & control
- Caliciviridae Infections/immunology
- Toll-Like Receptor 9/immunology
- Antibodies, Neutralizing/immunology
- Mice, Inbred C57BL
- Adjuvants, Vaccine
- Female
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Immunoglobulin G/immunology
- Immunity, Innate
- Humans
- Mice, Knockout
- CD8-Positive T-Lymphocytes/immunology
- Lipids/immunology
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/pharmacology
- Aluminum Hydroxide/immunology
- Liposomes
Collapse
Affiliation(s)
- Weiqian Dai
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Man Xing
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Lingjin Sun
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Lihui Lv
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Xiang Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yihan Wang
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Xueyang Pang
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Yingying Guo
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Jiling Ren
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
| | - Dongming Zhou
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, Tianjin, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Wang J, Zhao Z, Wang Q, Shi J, Wong DWC, Cheung JCW. Advancements in Nanoparticle-Based Adjuvants for Enhanced Tuberculosis Vaccination: A Review. Vaccines (Basel) 2024; 12:1335. [PMID: 39771997 PMCID: PMC11680411 DOI: 10.3390/vaccines12121335] [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: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective vaccines. Nanoparticle-based adjuvants represent a promising approach to enhancing tuberculosis vaccine efficacy. This review focuses on the advantages of nanoparticulate-loaded vaccines, emphasizing their ability to improve antigen delivery, safety, and immunogenicity. We discuss the various types of nanoparticles and their unique physicochemical properties that contribute to improved antigen delivery and sustained immune activation. Additionally, we highlight the advantages of nanoparticle-based adjuvants in inducing strong cellular and humoral immunity, enhancing vaccine stability, and reducing adverse effects. Finally, we address current challenges and future perspectives in the application of these novel adjuvants, emphasizing their potential to transform TB vaccine strategies and ultimately contribute to better global health outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Zian Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Quan Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Duo Wai-Chi Wong
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - James Chung-Wai Cheung
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
7
|
Catenacci L, Rossi R, Sechi F, Buonocore D, Sorrenti M, Perteghella S, Peviani M, Bonferoni MC. Effect of Lipid Nanoparticle Physico-Chemical Properties and Composition on Their Interaction with the Immune System. Pharmaceutics 2024; 16:1521. [PMID: 39771501 PMCID: PMC11728546 DOI: 10.3390/pharmaceutics16121521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid nanoparticles (LNPs) have shown promise as a delivery system for nucleic acid-based therapeutics, including DNA, siRNA, and mRNA vaccines. The immune system plays a critical role in the response to these nanocarriers, with innate immune cells initiating an early response and adaptive immune cells mediating a more specific reaction, sometimes leading to potential adverse effects. Recent studies have shown that the innate immune response to LNPs is mediated by Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs), which recognize the lipid components of the nanoparticles. This recognition can trigger the activation of inflammatory pathways and the production of cytokines and chemokines, leading to potential adverse effects such as fever, inflammation, and pain at the injection site. On the other hand, the adaptive immune response to LNPs appears to be primarily directed against the protein encoded by the mRNA cargo, with little evidence of an ongoing adaptive immune response to the components of the LNP itself. Understanding the relationship between LNPs and the immune system is critical for the development of safe and effective nucleic acid-based delivery systems. In fact, targeting the immune system is essential to develop effective vaccines, as well as therapies against cancer or infections. There is a lack of research in the literature that has systematically studied the factors that influence the interaction between LNPs and the immune system and further research is needed to better elucidate the mechanisms underlying the immune response to LNPs. In this review, we discuss LNPs' composition, physico-chemical properties, such as size, shape, and surface charge, and the protein corona formation which can affect the reactivity of the immune system, thus providing a guide for the research on new formulations that could gain a favorable efficacy/safety profile.
Collapse
Affiliation(s)
- Laura Catenacci
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Rachele Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Francesca Sechi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Daniela Buonocore
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Milena Sorrenti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Marco Peviani
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Maria Cristina Bonferoni
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| |
Collapse
|
8
|
Zhang R, Rygelski BT, Kruse LE, Smith JD, Wang X, Allen BN, Kramer JS, Seim GF, Faulkner TJ, Kuang H, Kokkoli E, Schrum AG, Ulery BD. Adjuvant Delivery Method and Nanoparticle Charge Influence Peptide Amphiphile Micelle Vaccine Bioactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598369. [PMID: 38915689 PMCID: PMC11195052 DOI: 10.1101/2024.06.10.598369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Vaccines are an indispensable public health measure that have enabled the eradication, near elimination, and prevention of a variety of pathogens. As research continues and our understanding of immunization strategies develops, subunit vaccines have emerged as exciting alternatives to existing whole vaccine approaches. Unfortunately, subunit vaccines often possess weak antigenicity, requiring delivery devices and adjuvant supplementation to improve their utility. Peptide amphiphile micelles have recently been shown to function as both delivery devices and self-adjuvanting systems that can be readily associated with molecular adjuvants to further improve vaccine-mediated host immunity. While promising, many design rules associated with the plethora of underlying adjustable parameters in the generation of a peptide amphiphile micelle vaccine have yet to be uncovered. This work explores the impact micellar adjuvant complexation method and incorporated antigen type have on their ability to activate dendritic cells and induce antigen specific responses. Interestingly, electrostatic complexation of CpG to micelles resulted in improved in vitro dendritic cell activation over hydrophobic association and antigen|adjuvant co-localization influenced cell-mediated, but not antibody-mediated immune responses. These exciting results complement those previously published to build the framework of a micelle vaccine toolbox that can be leveraged for future disease specific formulations.
Collapse
|
9
|
Li J, Foged C. Evaluating the breadth of nucleic acid-based payloads delivered in lipid nanoparticles to establish fundamental differences in development. Expert Opin Drug Deliv 2024; 21:1441-1461. [PMID: 39387233 DOI: 10.1080/17425247.2024.2409142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Nucleic acid (NA)-based therapeutics have shown great potential for downregulating or augmenting gene expression, and for promising applications, e.g., protein-replacement therapy and vaccination, a comprehensive understanding of the requirements for their targeted delivery to specific tissues or cells is needed. AREAS COVERED In this review, we discuss clinical applications of four representative types of NA-based therapeutics, i.e. antisense oligonucleotides, small interfering RNA, messenger RNA, and circular RNA, with a focus on the lipid nanoparticle (LNP) technology used for intracellular delivery. The in vivo fate of LNPs is discussed to improve the understanding of trafficking of nanomedicines at the systemic and cellular levels. In addition, NA-based vaccines are discussed, focusing on targeting antigen-presenting cells and immune activation. EXPERT OPINION Optimization of delivery systems for NA-based therapeutics is mainly focused on the standard requirements of prolonged systemic circulation and enhancing endosomal escape. Depending on the final destination in specific target tissues or cells, strategies should be adjusted to achieve the desired biodistribution of NA-based payloads. More studies relating to the pharmacokinetics of both cargo and carrier are encouraged, because their in vivo fates may differ, considering the possibility of premature cargo release before reaching the target.
Collapse
Affiliation(s)
- Jinjin Li
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| |
Collapse
|
10
|
Kang X, Mita N, Zhou L, Wu S, Yue Z, Babu RJ, Chen P. Nanotechnology in Advancing Chimeric Antigen Receptor T Cell Therapy for Cancer Treatment. Pharmaceutics 2024; 16:1228. [PMID: 39339264 PMCID: PMC11435308 DOI: 10.3390/pharmaceutics16091228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as a groundbreaking treatment for hematological cancers, yet it faces significant hurdles, particularly regarding its efficacy in solid tumors and concerning associated adverse effects. This review provides a comprehensive analysis of the advancements and ongoing challenges in CAR-T therapy. We highlight the transformative potential of nanotechnology in enhancing CAR-T therapy by improving targeting precision, modulating the immune-suppressive tumor microenvironment, and overcoming physical barriers. Nanotechnology facilitates efficient CAR gene delivery into T cells, boosting transfection efficiency and potentially reducing therapy costs. Moreover, nanotechnology offers innovative solutions to mitigate cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Cutting-edge nanotechnology platforms for real-time monitoring of CAR-T cell activity and cytokine release are also discussed. By integrating these advancements, we aim to provide valuable insights and pave the way for the next generation of CAR-T cell therapies to overcome current limitations and enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Xuejia Kang
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
| | - Nur Mita
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
- Faculty of Pharmacy, Mulawarman University, Samarinda 75119, Kalimantan Timur, Indonesia
| | - Lang Zhou
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
| | - Siqi Wu
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
| | - Zongliang Yue
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
| | - Pengyu Chen
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
| |
Collapse
|
11
|
Kim S, Jeon JH, Kim M, Lee Y, Hwang YH, Park M, Li CH, Lee T, Lee JA, Kim YM, Kim D, Lee H, Kim YJ, Kim VN, Park JE, Yeo J. Innate immune responses against mRNA vaccine promote cellular immunity through IFN-β at the injection site. Nat Commun 2024; 15:7226. [PMID: 39191748 PMCID: PMC11349762 DOI: 10.1038/s41467-024-51411-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
mRNA vaccines against SARS-CoV-2 have revolutionized vaccine development, but their immunological mechanisms are not fully understood. Here, we investigate injection site responses of mRNA vaccines by generating a comprehensive single-cell transcriptome profile upon lipid nanoparticle (LNP) or LNP-mRNA challenge in female BALB/c mice. We show that LNP-induced stromal pro-inflammatory responses and mRNA-elicited type I interferon responses dominate the initial injection site responses. By tracking the fate of delivered mRNA, we discover that injection site fibroblasts are highly enriched with the delivered mRNA and that they express IFN-β specifically in response to the mRNA component, not to the LNP component of mRNA vaccines. Moreover, the mRNA-LNP, but not LNP alone, induces migratory dendritic cells highly expressing IFN-stimulated genes (mDC_ISGs) at the injection site and draining lymph nodes. When co-injected with LNP-subunit vaccine, IFN-β induces mDC_ISGs at the injection site, and importantly, it substantially enhances antigen-specific cellular immune responses. Furthermore, blocking IFN-β signaling at the injection site significantly decreases mRNA vaccine-induced cellular immune responses. Collectively, these data highlight the importance of injection site fibroblasts and IFN-β signaling during early immune responses against the mRNA vaccine and provide detailed information on the initial chain of immune reactions elicited by mRNA vaccine injection.
Collapse
Affiliation(s)
- Seongryong Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ji Hyang Jeon
- Division of Infectious Disease Vaccine Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Republic of Korea
| | - Myeonghwan Kim
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University; Seodaemun-gu, Seoul, Republic of Korea
| | - Yun-Ho Hwang
- Division of Infectious Disease Vaccine Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Republic of Korea
| | - Myungsun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - C Han Li
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Taeyoung Lee
- Division of Infectious Disease Vaccine Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Republic of Korea
| | - Jung-Ah Lee
- Division of Infectious Disease Vaccine Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Republic of Korea
| | - You-Me Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Dokeun Kim
- Division of Infectious Disease Vaccine Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Republic of Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University; Seodaemun-gu, Seoul, Republic of Korea
| | - You-Jin Kim
- Division of Infectious Disease Vaccine Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Republic of Korea
| | - V Narry Kim
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| | - Jinah Yeo
- Division of Infectious Disease Vaccine Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Republic of Korea.
| |
Collapse
|
12
|
Barbieri BD, Peeler DJ, Samnuan K, Day S, Hu K, Sallah HJ, Tregoning JS, McKay PF, Shattock RJ. The role of helper lipids in optimising nanoparticle formulations of self-amplifying RNA. J Control Release 2024; 374:280-292. [PMID: 39142355 DOI: 10.1016/j.jconrel.2024.08.016] [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: 05/07/2024] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Lipid nanoparticle (LNP) formulation plays a vital role in RNA vaccine delivery. However, further optimisation of self-amplifying RNA (saRNA) vaccine formulation could help enhance seroconversion rates in humans and improve storage stability. Altering either the ionisable or helper lipid can alter the characteristics and performance of formulated saRNA through the interplay of the phospholipid's packing parameter and the geometrical shape within the LNP membrane. In this study, we compared the impact of three helper lipids (DSPC, DOPC, or DOPE) used with two different ionisable lipids (MC3 and C12-200) on stability, transfection efficiency and the inflammation and immunogenicity of saRNA. While helper lipid identity altered saRNA expression across four cell lines in vitro, this was not predictive of an ex vivo or in vivo response. The helper lipid used influenced LNP storage where DSPC provided the best stability profile over four weeks at 2-8 °C. Importantly, helper lipid impact on LNP storage stability was the best predictor of expression in human skin explants, where C12-200 in combination with DSPC provided the most durable expression. C12-200 LNPs also improved protein expression (firefly luciferase) and humoral responses to a SARS-CoV-2 spike saRNA vaccine compared to MC3 LNPs, where the effect of helper lipids was less apparent. Nevertheless, the performance of C12-200 in combination with DSPC appears optimal for saRNA when balancing preferred storage stability requirements against in vivo and ex vivo potency. These data suggest that helper lipid influences the stability and functionality of ionisable lipid nanoparticle-formulated saRNA.
Collapse
Affiliation(s)
| | - David J Peeler
- Department of Infectious Disease, Imperial College London, London, UK; Department of Materials, Imperial College London, London, UK
| | - Karnyart Samnuan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Suzanne Day
- Department of Infectious Disease, Imperial College London, London, UK
| | - Kai Hu
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Paul F McKay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
13
|
Fazel F, Doost JS, Raj S, Boodhoo N, Karimi K, Sharif S. The mRNA vaccine platform for veterinary species. Vet Immunol Immunopathol 2024; 274:110803. [PMID: 39003921 DOI: 10.1016/j.vetimm.2024.110803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Vaccination has proven to be an effective means of controlling pathogens in animals. Since the introduction of veterinary vaccines in the 19th century, several generations of vaccines have been introduced. These vaccines have had a positive impact on global animal health and production. Despite, the success of veterinary vaccines, there are still some pathogens for which there are no effective vaccines available, such as African swine fever. Further, animal health is under the constant threat of emerging and re-emerging pathogens, some of which are zoonotic and can pose a threat to human health. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the need for new vaccine platforms that are safe and efficacious, but also importantly, are adaptable and can be modified rapidly to match the circulating pathogens. mRNA vaccines have been shown to be an effective vaccine platform against various viral and bacterial pathogens. This review will cover some of the recent advances in the field of mRNA vaccines for veterinary species. Moreover, various mRNA vaccines and their delivery methods, as well as their reported efficacy, will be discussed. Current limitations and future prospects of this vaccine platform in veterinary medicine will also be discussed.
Collapse
Affiliation(s)
- Fatemeh Fazel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Janan Shoja Doost
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Sugandha Raj
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Nitish Boodhoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
14
|
Quan Y, Liu C, Lu X, Kong X, Yang S, Kong J, Wan W, Wang K, Xu K, Peng L. Comparison of the Immunogenicity of the LZ901 Vaccine and HZ/su Vaccine in a Mouse Model. Vaccines (Basel) 2024; 12:775. [PMID: 39066412 PMCID: PMC11281325 DOI: 10.3390/vaccines12070775] [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: 06/19/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Herpes zoster (HZ) is an infectious disease caused by the reactivation of varicella zoster virus (VZV), with 68% of cases occurring in adults over 50 years of age. HZ/su (Shingrix®) was approved by the Food and Drug Administration in 2017 for the prevention of HZ in individuals ≥ 50 years of age and showed very good protection from HZ. However, due to the use of the adjuvant AS01B, adverse reactions caused by Shingrix are a concern. Aluminum hydroxide is the most commonly used adjuvant and is widely used in a variety of vaccines. We developed a recombinant zoster vaccine (code: LZ901) consisting of a tetramer of VZV glycoprotein E (gE) and a human Fc fusion protein expressed in CHO cells, an immune complex-like molecule that can be adsorbed with an aluminum hydroxide adjuvant. We compared the immunogenicity of LZ901 with that of HZ/su in BALB/c mice. The results showed that LZ901 induced levels of gE-specific IgG antibodies comparable to those induced by HZ/su, and the results of FAMA titers further demonstrated their similar neutralizing antibody abilities. Most importantly, LZ901 induced higher levels of cell-mediated immunity (CMI) (which plays a decisive role in the efficacy of zoster vaccines) than HZ/su in BALB/c mice. The numbers of cytokine-producing T cells in LZ901-vaccinated mice were significantly greater than those in v-vaccinated mice, and the proportions of CD4+ and CD8+ T cells producing at least two types of cytokines in LZ901-vaccinated mice were significantly greater than those in HZ/su-vaccinated mice.
Collapse
Affiliation(s)
- Yaru Quan
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing 102629, China; (Y.Q.)
| | - Chunxia Liu
- Beijing Luzhu Biotechnology Co., Ltd., Beijing 101100, China
| | - Xu Lu
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing 102629, China; (Y.Q.)
| | - Xi Kong
- Beijing Luzhu Biotechnology Co., Ltd., Beijing 101100, China
| | - Shuai Yang
- Beijing Luzhu Biotechnology Co., Ltd., Beijing 101100, China
| | - Jian Kong
- Beijing Luzhu Biotechnology Co., Ltd., Beijing 101100, China
| | - Wenyan Wan
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing 102629, China; (Y.Q.)
| | - Kaiqin Wang
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing 102629, China; (Y.Q.)
| | - Kangwei Xu
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing 102629, China; (Y.Q.)
| | - Ling Peng
- Beijing Luzhu Biotechnology Co., Ltd., Beijing 101100, China
| |
Collapse
|
15
|
Kawai A, Noda M, Hirata H, Munakata L, Matsuda T, Omata D, Takemura N, Onoe S, Hirose M, Kato T, Saitoh T, Hirai T, Suzuki R, Yoshioka Y. Lipid Nanoparticle with 1,2-Di-O-octadecenyl-3-trimethylammonium-propane as a Component Lipid Confers Potent Responses of Th1 Cells and Antibody against Vaccine Antigen. ACS NANO 2024; 18:16589-16609. [PMID: 38885198 PMCID: PMC11223497 DOI: 10.1021/acsnano.4c00278] [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: 01/07/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024]
Abstract
Adjuvants are effective tools to enhance vaccine efficacy and control the type of immune responses such as antibody and T helper 1 (Th1)- or Th2-type responses. Several studies suggest that interferon (IFN)-γ-producing Th1 cells play a significant role against infections caused by intracellular bacteria and viruses; however, only a few adjuvants can induce a strong Th1-type immune response. Recently, several studies have shown that lipid nanoparticles (LNPs) can be used as vaccine adjuvants and that each LNP has a different adjuvant activity. In this study, we screened LNPs to develop an adjuvant that can induce Th1 cells and antibodies using a conventional influenza split vaccine (SV) as an antigen in mice. We observed that LNP with 1,2-di-O-octadecenyl-3-trimethylammonium-propane (DOTMA) as a component lipid (DOTMA-LNP) elicited robust SV-specific IgG1 and IgG2 responses compared with SV alone in mice and was as efficient as SV adjuvanted with other adjuvants in mice. Furthermore, DOTMA-LNPs induced robust IFN-γ-producing Th1 cells without inflammatory responses compared to those of other adjuvants, which conferred strong cross-protection in mice. We also demonstrated the high versatility of DOTMA-LNP as a Th1 cell-inducing vaccine adjuvant using vaccine antigens derived from severe acute respiratory syndrome coronavirus 2 and Streptococcus pneumoniae. Our findings suggest the potential of DOTMA-LNP as a safe and effective Th1 cell-inducing adjuvant and show that LNP formulations are potentially potent adjuvants to enhance the effectiveness of other subunit vaccines.
Collapse
Affiliation(s)
- Atsushi Kawai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Noda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Haruki Hirata
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Lisa Munakata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Teppei Matsuda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Daiki Omata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Naoki Takemura
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sakura Onoe
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mika Hirose
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kato
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tatsuya Saitoh
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshiro Hirai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Suzuki
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Yasuo Yoshioka
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, The Research Foundation for Microbial Diseases of
Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
16
|
Saleemi MA, Zhang Y, Zhang G. Current Progress in the Science of Novel Adjuvant Nano-Vaccine-Induced Protective Immune Responses. Pathogens 2024; 13:441. [PMID: 38921739 PMCID: PMC11206999 DOI: 10.3390/pathogens13060441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Vaccinations are vital as they protect us from various illness-causing agents. Despite all the advancements in vaccine-related research, developing improved and safer vaccines against devastating infectious diseases including Ebola, tuberculosis and acquired immune deficiency syndrome (AIDS) remains a significant challenge. In addition, some of the current human vaccines can cause adverse reactions in some individuals, which limits their use for massive vaccination program. Therefore, it is necessary to design optimal vaccine candidates that can elicit appropriate immune responses but do not induce side effects. Subunit vaccines are relatively safe for the vaccination of humans, but they are unable to trigger an optimal protective immune response without an adjuvant. Although different types of adjuvants have been used for the formulation of vaccines to fight pathogens that have high antigenic diversity, due to the toxicity and safety issues associated with human-specific adjuvants, there are only a few adjuvants that have been approved for the formulation of human vaccines. Recently, nanoparticles (NPs) have gain specific attention and are commonly used as adjuvants for vaccine development as well as for drug delivery due to their excellent immune modulation properties. This review will focus on the current state of adjuvants in vaccine development, the mechanisms of human-compatible adjuvants and future research directions. We hope this review will provide valuable information to discovery novel adjuvants and drug delivery systems for developing novel vaccines and treatments.
Collapse
Affiliation(s)
| | | | - Guoquan Zhang
- Department of Molecular Microbiology and Immunology, College of Sciences, University of Texas at San Antonio, San Antonio, TX 78249, USA; (M.A.S.); (Y.Z.)
| |
Collapse
|
17
|
Bonam SR, Hazell NC, Mathew MJ, Liang Y, Zhang X, Wei Z, Alameh MG, Weissman D, Hu H. Innate and Adaptive Immune Parameters following mRNA Vaccination in Mice. Vaccines (Basel) 2024; 12:543. [PMID: 38793794 PMCID: PMC11125668 DOI: 10.3390/vaccines12050543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
The COVID-19 pandemic has raised the standard regarding the current vaccine development pace, as several messenger RNA (mRNA)-lipid nanoparticle (LNP) vaccines have proved their ability to induce strong immunogenicity and protective efficacy. We developed 1-methylpseudouridine-containing mRNA-LNP vaccines, expressing either the more conserved SARS-CoV-2 nucleoprotein (mRNA-N) or spike protein (mRNA-S), both based on the prototypic viral sequences. When combining both mRNA-S and mRNA-N together (mRNA-S+N), the vaccine showed high immunogenicity and broad protection against different SARS-CoV-2 variants, including wildtype, Delta, BA.1, BA.5, and BQ.1. To better understand the mechanisms behind this broad protection obtained by mRNA-S+N, we analyzed innate and adaptive immune parameters following vaccination in mice. Compared to either mRNA-S or mRNA-N alone, mice vaccinated with mRNA-S+N exhibited an increase in the innate immune response, as depicted by the higher cytokine (IL-6 and chemokine (MCP-1) levels. In addition, lymph node immunophenotyping showed the maturation and activation of dendritic cells and natural killer cells, respectively. To understand the adaptive immune response, RNA-Seq analyses of the lung and spleen samples of the vaccinated mice were performed in parallel and revealed a stronger immune gene-expression profile in the lung than that in the spleen. Compared to mRNA-S alone, mRNA-S+N vaccination elicited higher levels of expression for genes involved in multiple immune pathways, including T cells, cytokine signaling, antigen presentation, B cells, and innate immunity. Together, our studies provide immunological insights into the mechanisms of broad protection conferred by dual mRNA vaccination against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
| | - Nicholas C. Hazell
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mano Joseph Mathew
- EFREI Research Lab, Panthéon Assas University, 30-32 Avenue de la République, 94800 Villejuif, France;
- Laboratoire Génomique, Bioinformatique et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, 2 rue Conté, 75003 Paris, France
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
| | - Xuxiang Zhang
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA; (X.Z.); (Z.W.)
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA; (X.Z.); (Z.W.)
| | - Mohamad-Gabriel Alameh
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA; (M.-G.A.); (D.W.)
| | - Drew Weissman
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA; (M.-G.A.); (D.W.)
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
18
|
Omo-Lamai S, Wang Y, Patel MN, Essien EO, Shen M, Majumdar A, Espy C, Wu J, Channer B, Tobin M, Murali S, Papp TE, Maheshwari R, Wang L, Chase LS, Zamora ME, Arral ML, Marcos-Contreras OA, Myerson JW, Hunter CA, Tsourkas A, Muzykantov V, Brodsky I, Shin S, Whitehead KA, Gaskill P, Discher D, Parhiz H, Brenner JS. Lipid Nanoparticle-Associated Inflammation is Triggered by Sensing of Endosomal Damage: Engineering Endosomal Escape Without Side Effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589801. [PMID: 38659905 PMCID: PMC11042321 DOI: 10.1101/2024.04.16.589801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as the dominant platform for RNA delivery, based on their success in the COVID-19 vaccines and late-stage clinical studies in other indications. However, we and others have shown that LNPs induce severe inflammation, and massively aggravate pre-existing inflammation. Here, using structure-function screening of lipids and analyses of signaling pathways, we elucidate the mechanisms of LNP-associated inflammation and demonstrate solutions. We show that LNPs' hallmark feature, endosomal escape, which is necessary for RNA expression, also directly triggers inflammation by causing endosomal membrane damage. Large, irreparable, endosomal holes are recognized by cytosolic proteins called galectins, which bind to sugars on the inner endosomal membrane and then regulate downstream inflammation. We find that inhibition of galectins abrogates LNP-associated inflammation, both in vitro and in vivo . We show that rapidly biodegradable ionizable lipids can preferentially create endosomal holes that are smaller in size and reparable by the endosomal sorting complex required for transport (ESCRT) pathway. Ionizable lipids producing such ESCRT-recruiting endosomal holes can produce high expression from cargo mRNA with minimal inflammation. Finally, we show that both routes to non-inflammatory LNPs, either galectin inhibition or ESCRT-recruiting ionizable lipids, are compatible with therapeutic mRNAs that ameliorate inflammation in disease models. LNPs without galectin inhibition or biodegradable ionizable lipids lead to severe exacerbation of inflammation in these models. In summary, endosomal escape induces endosomal membrane damage that can lead to inflammation. However, the inflammation can be controlled by inhibiting galectins (large hole detectors) or by using biodegradable lipids, which create smaller holes that are reparable by the ESCRT pathway. These strategies should lead to generally safer LNPs that can be used to treat inflammatory diseases.
Collapse
|
19
|
Wu N, Zhang J, Shen Y, Zhang X, Zhou J, Wu Y, Li E, Meng X, Chuai X, Chiu S, Wang Y. A potential bivalent mRNA vaccine candidate protects against both RSV and SARS-CoV-2 infections. Mol Ther 2024; 32:1033-1047. [PMID: 38341613 PMCID: PMC11163217 DOI: 10.1016/j.ymthe.2024.02.011] [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: 08/31/2023] [Revised: 12/29/2023] [Accepted: 02/07/2024] [Indexed: 02/12/2024] Open
Abstract
As the world continues to confront severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), respiratory syncytial virus (RSV) is also causing severe respiratory illness in millions of infants, elderly individuals, and immunocompromised people globally. Exacerbating the situation is the fact that co-infection with multiple viruses is occurring, something which has greatly increased the clinical severity of the infections. Thus, our team developed a bivalent vaccine that delivered mRNAs encoding SARS-CoV-2 Omicron spike (S) and RSV fusion (F) proteins simultaneously, SF-LNP, which induced S and F protein-specific binding antibodies and cellular immune responses in BALB/c mice. Moreover, SF-LNP immunization effectively protected BALB/c mice from RSV infection and hamsters from SARS-CoV-2 Omicron infection. Notably, our study pointed out the antigenic competition problem of bivalent vaccines and provided a solution. Overall, our results demonstrated the potential of preventing two infectious diseases with a single vaccine and provided a paradigm for the subsequent design of multivalent vaccines.
Collapse
Affiliation(s)
- Namei Wu
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, P.R. China; School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Jiachen Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Yanqiong Shen
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Xinghai Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China
| | - Jinge Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China
| | - Yan Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China
| | - Entao Li
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei 230027, P.R. China
| | - Xia Chuai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430062, P.R. China.
| | - Sandra Chiu
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China.
| | - Yucai Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, P.R. China; School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, P.R. China; RNAlfa Biotech, Hefei 230088, P.R. China.
| |
Collapse
|
20
|
Sun Z, Zhao H, Ma L, Shi Y, Ji M, Sun X, Ma D, Zhou W, Huang T, Zhang D. The quest for nanoparticle-powered vaccines in cancer immunotherapy. J Nanobiotechnology 2024; 22:61. [PMID: 38355548 PMCID: PMC10865557 DOI: 10.1186/s12951-024-02311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Despite recent advancements in cancer treatment, this disease still poses a serious threat to public health. Vaccines play an important role in preventing illness by preparing the body's adaptive and innate immune responses to combat diseases. As our understanding of malignancies and their connection to the immune system improves, there has been a growing interest in priming the immune system to fight malignancies more effectively and comprehensively. One promising approach involves utilizing nanoparticle systems for antigen delivery, which has been shown to potentiate immune responses as vaccines and/or adjuvants. In this review, we comprehensively summarized the immunological mechanisms of cancer vaccines while focusing specifically on the recent applications of various types of nanoparticles in the field of cancer immunotherapy. By exploring these recent breakthroughs, we hope to identify significant challenges and obstacles in making nanoparticle-based vaccines and adjuvants feasible for clinical application. This review serves to assess recent breakthroughs in nanoparticle-based cancer vaccinations and shed light on their prospects and potential barriers. By doing so, we aim to inspire future immunotherapies for cancer that harness the potential of nanotechnology to deliver more effective and targeted treatments.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hui Zhao
- Department of Endodontics, East Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Li Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanli Shi
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mei Ji
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Dan Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wei Zhou
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tao Huang
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Dongsheng Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
21
|
Sharma P, Hoorn D, Aitha A, Breier D, Peer D. The immunostimulatory nature of mRNA lipid nanoparticles. Adv Drug Deliv Rev 2024; 205:115175. [PMID: 38218350 DOI: 10.1016/j.addr.2023.115175] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024]
Abstract
mRNA-Lipid nanoparticles (LNPs) are at the forefront of global medical research. With the development of mRNA-LNP vaccines to combat the COVID-19 pandemic, the clinical potential of this platform was unleashed. Upon administering 16 billion doses that protected billions of people, it became clear that a fraction of them witnessed mild and in some cases even severe adverse effects. Therefore, it is paramount to define the safety along with the therapeutic efficacy of the mRNA-LNP platform for the successful translation of new genetic medicines based on this technology. While mRNA was the effector molecule of this platform, the ionizable lipid component of the LNPs played an indispensable role in its success. However, both of these components possess the ability to induce undesired immunostimulation, which is an area that needs to be addressed systematically. The immune cell agitation caused by this platform is a two-edged sword as it may prove beneficial for vaccination but detrimental to other applications. Therefore, a key challenge in advancing the mRNA-LNP drug delivery platform from bench to bedside is understanding the immunostimulatory behavior of these components. Herein, we provide a detailed overview of the structural modifications and immunogenicity of synthetic mRNA. We discuss the effect of ionizable lipid structure on LNP functionality and offer a mechanistic overview of the ability of LNPs to elicit an immune response. Finally, we shed some light on the current status of this technology in clinical trials and discuss a few challenges to be addressed to advance the field.
Collapse
Affiliation(s)
- Preeti Sharma
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Daniek Hoorn
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Anjaiah Aitha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dor Breier
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
22
|
Anindita J, Tanaka H, Yamakawa T, Sato Y, Matsumoto C, Ishizaki K, Oyama T, Suzuki S, Ueda K, Higashi K, Moribe K, Sasaki K, Ogura Y, Yonemochi E, Sakurai Y, Hatakeyama H, Akita H. The Effect of Cholesterol Content on the Adjuvant Activity of Nucleic-Acid-Free Lipid Nanoparticles. Pharmaceutics 2024; 16:181. [PMID: 38399242 PMCID: PMC10893020 DOI: 10.3390/pharmaceutics16020181] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
RNA vaccines are applicable to the treatment of various infectious diseases via the inducement of robust immune responses against target antigens by expressing antigen proteins in the human body. The delivery of messenger RNA by lipid nanoparticles (LNPs) has become a versatile drug delivery system used in the administration of RNA vaccines. LNPs are widely considered to possess adjuvant activity that induces a strong immune response. However, the properties of LNPs that contribute to their adjuvant activity continue to require clarification. To characterize the relationships between the lipid composition, particle morphology, and adjuvant activity of LNPs, the nanostructures of LNPs and their antibody production were evaluated. To simply compare the adjuvant activity of LNPs, empty LNPs were subcutaneously injected with recombinant proteins. Consistent with previous research, the presence of ionizable lipids was one of the determinant factors. Adjuvant activity was induced when a tiny cholesterol assembly (cholesterol-induced phase, ChiP) was formed according to the amount of cholesterol present. Moreover, adjuvant activity was diminished when the content of cholesterol was excessive. Thus, it is plausible that an intermediate structure of cholesterol (not in a crystalline-like state) in an intra-particle space could be closely related to the immunogenicity of LNPs.
Collapse
Affiliation(s)
- Jessica Anindita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba Aramaki, Aoba-ku, Sendai City 980-8578, Miyagi, Japan
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba Aramaki, Aoba-ku, Sendai City 980-8578, Miyagi, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita 565-0871, Osaka, Japan
| | - Takuma Yamakawa
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan
| | - Yuka Sato
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan
| | - Chika Matsumoto
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba Aramaki, Aoba-ku, Sendai City 980-8578, Miyagi, Japan
| | - Kota Ishizaki
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan
| | - Taiji Oyama
- Sales Division, JASCO Corporation, 2967-5 Ishikawa, Hachioji City 192-8537, Tokyo, Japan;
| | - Satoko Suzuki
- Applicative Solution Lab Division, JASCO Corporation, 2967-5 Ishikawa, Hachioji City 192-8537, Tokyo, Japan
| | - Keisuke Ueda
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan; (K.U.)
| | - Kenjirou Higashi
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan; (K.U.)
| | - Kunikazu Moribe
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan; (K.U.)
| | - Kasumi Sasaki
- Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Shinagawa City 142-8501, Tokyo, Japan
| | - Yumika Ogura
- Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Shinagawa City 142-8501, Tokyo, Japan
| | - Etsuo Yonemochi
- Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Shinagawa City 142-8501, Tokyo, Japan
| | - Yu Sakurai
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba Aramaki, Aoba-ku, Sendai City 980-8578, Miyagi, Japan
| | - Hiroto Hatakeyama
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-0856, Chiba, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba Aramaki, Aoba-ku, Sendai City 980-8578, Miyagi, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita 565-0871, Osaka, Japan
| |
Collapse
|
23
|
Xie C, Yao R, Xia X. The advances of adjuvants in mRNA vaccines. NPJ Vaccines 2023; 8:162. [PMID: 37884526 PMCID: PMC10603121 DOI: 10.1038/s41541-023-00760-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
The remarkable success of messenger RNA (mRNA) vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has propelled the rapid development of this vaccination technology in recent years. Over the last three decades, numerous studies have shown the considerable potential of mRNA vaccines that elicit protective immune responses against pathogens or cancers in preclinical studies or clinical trials. These effective mRNA vaccines usually contain specific adjuvants to obtain the desired immune effect. Vaccine adjuvants traditionally are immunopotentiators that bind to pattern recognition receptors (PRRs) of innate immune cells to increase the magnitude or achieve qualitative alteration of immune responses, finally enhancing the efficacy of vaccines. Generally, adjuvants are necessary parts of competent vaccines. According to the existing literature, adjuvants in mRNA vaccines can be broadly classified into three categories: 1) RNA with self-adjuvant characteristics, 2) components of the delivery system, and 3) exogenous immunostimulants. This review summarizes the three types of adjuvants used in mRNA vaccines and provides a comprehensive understanding of molecular mechanisms by which adjuvants exert their functions in mRNA vaccines.
Collapse
Affiliation(s)
- Chunyuan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ruhui Yao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
24
|
Han X, Alameh MG, Butowska K, Knox JJ, Lundgreen K, Ghattas M, Gong N, Xue L, Xu Y, Lavertu M, Bates P, Xu J, Nie G, Zhong Y, Weissman D, Mitchell MJ. Adjuvant lipidoid-substituted lipid nanoparticles augment the immunogenicity of SARS-CoV-2 mRNA vaccines. NATURE NANOTECHNOLOGY 2023; 18:1105-1114. [PMID: 37365276 DOI: 10.1038/s41565-023-01404-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 04/17/2023] [Indexed: 06/28/2023]
Abstract
Lipid nanoparticle (LNP)-formulated messenger RNA (mRNA) vaccineare a promising platform to prevent infectious diseases as demonstrated by the recent success of SARS-CoV-2 mRNA vaccines. To avoid immune recognition and uncontrolled inflammation, nucleoside-modified mRNA is used. However, such modification largely abrogates the innate immune responses that are critical to orchestrating robust adaptive immunity. Here we develop an LNP component-an adjuvant lipidoid-that can enhance the adjuvanticity of mRNA-LNP vaccines. Our results show that partial substitution of ionizable lipidoid with adjuvant lipidoid not only enhanced mRNA delivery, but also endowed LNPs with Toll-like receptor 7/8-agonistic activity, which significantly increased the innate immunity of the SARS-CoV-2 mRNA-LNP vaccine with good tolerability in mice. Our optimized vaccine elicits potent neutralizing antibodies against multiple SARS-CoV-2 pseudovirus variants, strong Th1-biased cellular immunity, and robust B cell and long-lived plasma cell responses. Importantly, this adjuvant lipidoid substitution strategy works successfully in a clinically relevant mRNA-LNP vaccine, demonstrating its translational potential.
Collapse
Affiliation(s)
- Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Kamila Butowska
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Intercollegiate Faculty of Biotechnology, University of Gdańsk & Medical University of Gdańsk, Gdańsk, Poland
| | - James J Knox
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendall Lundgreen
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Majed Ghattas
- Department of Chemical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Xu
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Marc Lavertu
- Department of Chemical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
| | - Paul Bates
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, People's Republic of China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, People's Republic of China
| | - Yi Zhong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Jeong M, Lee Y, Park J, Jung H, Lee H. Lipid nanoparticles (LNPs) for in vivo RNA delivery and their breakthrough technology for future applications. Adv Drug Deliv Rev 2023; 200:114990. [PMID: 37423563 DOI: 10.1016/j.addr.2023.114990] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
RNA therapeutics show a significant breakthrough for the treatment of otherwise incurable diseases and genetic disorders by regulating disease-related gene expression. The successful development of COVID-19 mRNA vaccines further emphasizes the potential of RNA therapeutics in the prevention of infectious diseases as well as in the treatment of chronic diseases. However, the efficient delivery of RNA into cells remains a challenge, and nanoparticle delivery systems such as lipid nanoparticles (LNPs) are necessary to fully realize the potential of RNA therapeutics. While LNPs provide a highly efficient platform for the in vivo delivery of RNA by overcoming various biological barriers, several challenges remain to be resolved for further development and regulatory approval. These include a lack of targeted delivery to extrahepatic organs and a gradual loss of therapeutic potency with repeated doses. In this review, we highlight the fundamental aspects of LNPs and their uses in the development of novel RNA therapeutics. Recent advances in LNP-based therapeutics and preclinical/clinical studies are overviewed. Lastly, we discuss the current limitations of LNPs and introduce breakthrough technologies that might overcome these challenges in future applications.
Collapse
Affiliation(s)
- Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Yeji Lee
- 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
|
Huang Z, Zhang Y, Li H, Zhu J, Song W, Chen K, Zhang Y, Lou Y. Vaccine development for mosquito-borne viral diseases. Front Immunol 2023; 14:1161149. [PMID: 37251387 PMCID: PMC10213220 DOI: 10.3389/fimmu.2023.1161149] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
Mosquito-borne viral diseases are a group of viral illnesses that are predominantly transmitted by mosquitoes, including viruses from the Togaviridae and Flaviviridae families. In recent years, outbreaks caused by Dengue and Zika viruses from the Flaviviridae family, and Chikungunya virus from the Togaviridae family, have raised significant concerns for public health. However, there are currently no safe and effective vaccines available for these viruses, except for CYD-TDV, which has been licensed for Dengue virus. Efforts to control the transmission of COVID-19, such as home quarantine and travel restrictions, have somewhat limited the spread of mosquito-borne viral diseases. Several vaccine platforms, including inactivated vaccines, viral-vector vaccines, live attenuated vaccines, protein vaccines, and nucleic acid vaccines, are being developed to combat these viruses. This review analyzes the various vaccine platforms against Dengue, Zika, and Chikungunya viruses and provides valuable insights for responding to potential outbreaks.
Collapse
Affiliation(s)
- Zhiwei Huang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuxuan Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hongyu Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiajie Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Wanchen Song
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yanjun Zhang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yongliang Lou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
27
|
Brito C, Lourenço C, Magalhães J, Reis S, Borges M. Nanoparticles as a Delivery System of Antigens for the Development of an Effective Vaccine against Toxoplasma gondii. Vaccines (Basel) 2023; 11:vaccines11040733. [PMID: 37112645 PMCID: PMC10142924 DOI: 10.3390/vaccines11040733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Nanoparticles include particles ranging in size from nanometers to micrometers, whose physicochemical characteristics are optimized to make them appropriate delivery vehicles for drugs or immunogens important in the fight and/or prevention of infectious diseases. There has been a rise in the use of nanoparticles in preventive vaccine formulations as immunostimulatory adjuvants, and as vehicles for immunogen delivery to target immune cells. Toxoplasma is important worldwide, and may cause human toxoplasmosis. In immunocompetent hosts, infection is usually asymptomatic, but in immunocompromised patients it can cause serious neurological and ocular consequences, such as encephalitis and retinochoroiditis. Primary infection during pregnancy may cause abortion or congenital toxoplasmosis. Currently, there is no effective human vaccine against this disease. Evidence has emerged from several experimental studies testing nanovaccines showing them to be promising tools in the prevention of experimental toxoplasmosis. For the present study, a literature review was carried out on articles published over the last 10 years through the PubMed database, pertaining to in vivo experimental models of T. gondii infection where nanovaccines were tested and protection and immune responses evaluated. This review aims to highlight the way forward in the search for an effective vaccine for toxoplasmosis.
Collapse
|
28
|
Lam AK, Roshan R, Miley W, Labo N, Zhen J, Kurland AP, Cheng C, Huang H, Teng PL, Harelson C, Gong D, Tam YK, Radu CG, Epeldegui M, Johnson JR, Zhou ZH, Whitby D, Wu TT. Immunization of Mice with Virus-Like Vesicles of Kaposi Sarcoma-Associated Herpesvirus Reveals a Role for Antibodies Targeting ORF4 in Activating Complement-Mediated Neutralization. J Virol 2023; 97:e0160022. [PMID: 36757205 PMCID: PMC9972917 DOI: 10.1128/jvi.01600-22] [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: 10/14/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
Infection by Kaposi sarcoma-associated herpesvirus (KSHV) can cause severe consequences, such as cancers and lymphoproliferative diseases. Whole inactivated viruses (WIV) with chemically destroyed genetic materials have been used as antigens in several licensed vaccines. During KSHV productive replication, virus-like vesicles (VLVs) that lack capsids and viral genomes are generated along with virions. Here, we investigated the immunogenicity of KSHV VLVs produced from a viral mutant that was defective in capsid formation and DNA packaging. Mice immunized with adjuvanted VLVs generated KSHV-specific T cell and antibody responses. Neutralization of KSHV infection by the VLV immune serum was low but was markedly enhanced in the presence of the complement system. Complement-enhanced neutralization and complement deposition on KSHV-infected cells was dependent on antibodies targeting viral open reading frame 4 (ORF4). However, limited complement-mediated enhancement was detected in the sera of a small cohort of KSHV-infected humans which contained few neutralizing antibodies. Therefore, vaccination that induces antibody effector functions can potentially improve infection-induced humoral immunity. Overall, our study highlights a potential benefit of engaging complement-mediated antibody functions in future KSHV vaccine development. IMPORTANCE KSHV is a virus that can lead to cancer after infection. A vaccine that prevents KSHV infection or transmission would be helpful in preventing the development of these cancers. We investigated KSHV VLV as an immunogen for vaccination. We determined that antibodies targeting the viral protein ORF4 induced by VLV immunization could engage the complement system and neutralize viral infection. However, ORF4-specific antibodies were seldom detected in the sera of KSHV-infected humans. Moreover, these human sera did not potently trigger complement-mediated neutralization, indicating an improvement that immunization can confer. Our study suggests a new antibody-mediated mechanism to control KSHV infection and underscores the benefit of activating the complement system in a future KSHV vaccine.
Collapse
Affiliation(s)
- Alex K. Lam
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Romin Roshan
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Wendell Miley
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Nazzarena Labo
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - James Zhen
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Andrew P. Kurland
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Celine Cheng
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Haigen Huang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Pu-Lin Teng
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Claire Harelson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Danyang Gong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, British Columbia, Canada
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Marta Epeldegui
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jeffrey R. Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Z. Hong Zhou
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Denise Whitby
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
29
|
Rohde CM, Lindemann C, Giovanelli M, Sellers RS, Diekmann J, Choudhary S, Ramaiah L, Vogel AB, Chervona Y, Muik A, Sahin U. Toxicological Assessments of a Pandemic COVID-19 Vaccine-Demonstrating the Suitability of a Platform Approach for mRNA Vaccines. Vaccines (Basel) 2023; 11:417. [PMID: 36851293 PMCID: PMC9965811 DOI: 10.3390/vaccines11020417] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
The emergence of SARS-CoV-2 at the end of 2019 required the swift development of a vaccine to address the pandemic. Nonclinical GLP-compliant studies in Wistar Han rats were initiated to assess the local tolerance, systemic toxicity, and immune response to four mRNA vaccine candidates encoding immunogens derived from the spike (S) glycoprotein of SARS-CoV-2, encapsulated in lipid nanoparticles (LNPs). Vaccine candidates were administered intramuscularly once weekly for three doses at 30 and/or 100 µg followed by a 3-week recovery period. Clinical pathology findings included higher white blood cell counts and acute phase reactant concentrations, lower platelet and reticulocyte counts, and lower RBC parameters. Microscopically, there was increased cellularity (lymphocytes) in the lymph nodes and spleen, increased hematopoiesis in the bone marrow and spleen, acute inflammation and edema at the injection site, and minimal hepatocellular vacuolation. These findings were generally attributed to the anticipated immune and inflammatory responses to the vaccines, except for hepatocyte vacuolation, which was interpreted to reflect hepatocyte LNP lipid uptake, was similar between candidates and resolved or partially recovered at the end of the recovery phase. These studies demonstrated safety and tolerability in rats, supporting SARS-CoV-2 mRNA-LNP vaccine clinical development.
Collapse
Affiliation(s)
- Cynthia M. Rohde
- Drug Safety Research and Development, Pfizer Worldwide Research, Development & Medical, Pfizer, Inc., Pearl River, NY 10965, USA
| | | | - Michael Giovanelli
- Drug Safety Research and Development, Pfizer Worldwide Research, Development & Medical, Pfizer, Inc., Groton, CT 06340, USA
| | - Rani S. Sellers
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Shambhunath Choudhary
- Drug Safety Research and Development, Pfizer Worldwide Research, Development & Medical, Pfizer, Inc., Pearl River, NY 10965, USA
| | - Lila Ramaiah
- Drug Safety Research and Development, Pfizer Worldwide Research, Development & Medical, Pfizer, Inc., Pearl River, NY 10965, USA
| | | | - Yana Chervona
- Drug Safety Research and Development, Pfizer Worldwide Research, Development & Medical, Pfizer, Inc., Pearl River, NY 10965, USA
| | | | | |
Collapse
|
30
|
García-Silva I, Govea-Alonso DO, Rosales-Mendoza S. Current status of mucosal vaccines against SARS-CoV2: a hope for protective immunity. Expert Opin Biol Ther 2023; 23:207-222. [PMID: 36594264 DOI: 10.1080/14712598.2022.2156284] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The current vaccines used to fight against COVID-19 are effective, however the induction of protective immunity is a pending goal required to prevent viral transmission, prevent the generation of new variants, and ultimately eradicate SARS-CoV-2. Mucosal immunization stands as a promising approach to achieve protective immunity against SARS-CoV-2; therefore, it is imperative to innovate the current vaccines by developing mucosal candidates, focusing not only on their ability to prevent severe COVID-19 but to neutralize the virus before invasion of the respiratory system and other mucosal compartments. AREAS COVERED This review covers the current advances on the development of anti-COVID-19 mucosal vaccines. Biomedical literature, including PubMed and clinicaltrials.gov website, was analyzed to identify the state of the art for this field. The achievements in preclinical and clinical evaluations are presented and critically analyzed. EXPERT OPINION There is a significant advance on the development of mucosal vaccines against SARSCoV-2, which is a promise to increase the efficacy of immunization against this pathogen. Both preclinical and clinical evaluation for several candidates have been performed. The challenges in this road (e.g. low immunogenicity, a reduced number of adjuvants available, and inaccurate dosage) are identified and also critical perspectives for the field are provided.
Collapse
Affiliation(s)
- Ileana García-Silva
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| | - Dania O Govea-Alonso
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| |
Collapse
|
31
|
Austin LA, Smith JS, Nahas DD, Danzinger A, Secore S, O'Donnell G, Radcliffe S, Hu S, Perley J, Bett AJ, Gindy ME. Split-Dose Administration Enhances Immune Responses Elicited by a mRNA/Lipid Nanoparticle Vaccine Expressing Respiratory Syncytial Virus F Protein. Mol Pharm 2023; 20:279-289. [PMID: 36251490 DOI: 10.1021/acs.molpharmaceut.2c00635] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
mRNA vaccines have recently received significant attention due to their role in combating the SARS-CoV-2 pandemic. As a platform, mRNA vaccines have been shown to elicit strong humoral and cellular immune responses with acceptable safety profiles for prophylactic use. Despite their potential, industrial challenges have limited realization of the vaccine platform on a global scale. Critical among these challenges are supply chain considerations, including mRNA production, cost of goods, and vaccine frozen-chain distribution. Here, we assess the delivery of lipid nanoparticle-encapsulated mRNA (mRNA/LNP) vaccines using a split-dose immunization regimen as an approach to develop mRNA dose-sparing vaccine regimens with potential to mitigate mRNA supply chain challenges. Our data demonstrate that immunization by a mRNA/LNP vaccine encoding respiratory syncytial virus pre-F (RSV pre-F) over a 9 day period elicits comparable or superior magnitude of antibodies when compared to traditional bolus immunization of the vaccine. The split-dose immunization regimens evaluated in our studies were designed to mimic reported drug or antigen release profiles from microneedle patches, highlighting the potential benefit of pairing mRNA vaccines with patch-based delivery technologies to enable sustained release and solid-state stabilization. Overall, our findings provide a proof of concept to support further investigations into the development of sustained delivery approaches for mRNA/LNP vaccines.
Collapse
Affiliation(s)
- Lauren A Austin
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | - Jeffrey S Smith
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | - Debbie D Nahas
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | | | - Susan Secore
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | | | - Scott Radcliffe
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | - Shuai Hu
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | - Jeffrey Perley
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | - Andrew J Bett
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| | - Marian E Gindy
- Merck & Co., Inc., Rahway, New Jersey 07033-1310, United States
| |
Collapse
|
32
|
Matarazzo L, Bettencourt PJG. mRNA vaccines: a new opportunity for malaria, tuberculosis and HIV. Front Immunol 2023; 14:1172691. [PMID: 37168860 PMCID: PMC10166207 DOI: 10.3389/fimmu.2023.1172691] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
The success of the first licensed mRNA-based vaccines against COVID-19 has created a widespread interest on mRNA technology for vaccinology. As expected, the number of mRNA vaccines in preclinical and clinical development increased exponentially since 2020, including numerous improvements in mRNA formulation design, delivery methods and manufacturing processes. However, the technology faces challenges such as the cost of raw materials, the lack of standardization, and delivery optimization. MRNA technology may provide a solution to some of the emerging infectious diseases as well as the deadliest hard-to-treat infectious diseases malaria, tuberculosis, and human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS), for which an effective vaccine, easily deployable to endemic areas is urgently needed. In this review, we discuss the functional structure, design, manufacturing processes and delivery methods of mRNA vaccines. We provide an up-to-date overview of the preclinical and clinical development of mRNA vaccines against infectious diseases, and discuss the immunogenicity, efficacy and correlates of protection of mRNA vaccines, with particular focus on research and development of mRNA vaccines against malaria, tuberculosis and HIV.
Collapse
Affiliation(s)
- Laura Matarazzo
- Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Lisboa, Portugal
- Faculty of Medicine, Universidade Católica Portuguesa, Rio de Mouro, Portugal
| | - Paulo J. G. Bettencourt
- Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Lisboa, Portugal
- Faculty of Medicine, Universidade Católica Portuguesa, Rio de Mouro, Portugal
- *Correspondence: Paulo J. G. Bettencourt,
| |
Collapse
|
33
|
Verbeke R, Hogan MJ, Loré K, Pardi N. Innate immune mechanisms of mRNA vaccines. Immunity 2022; 55:1993-2005. [PMID: 36351374 PMCID: PMC9641982 DOI: 10.1016/j.immuni.2022.10.014] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/08/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022]
Abstract
The lipid nanoparticle (LNP)-encapsulated, nucleoside-modified mRNA platform has been used to generate safe and effective vaccines in record time against COVID-19. Here, we review the current understanding of the manner whereby mRNA vaccines induce innate immune activation and how this contributes to protective immunity. We discuss innate immune sensing of mRNA vaccines at the cellular and intracellular levels and consider the contribution of both the mRNA and the LNP components to their immunogenicity. A key message that is emerging from recent observations is that the LNP carrier acts as a powerful adjuvant for this novel vaccine platform. In this context, we highlight important gaps in understanding and discuss how new insight into the mechanisms underlying the effectiveness of mRNA-LNP vaccines may enable tailoring mRNA and carrier molecules to develop vaccines with greater effectiveness and milder adverse events in the future.
Collapse
Affiliation(s)
- Rein Verbeke
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Department of Biochemistry and Molecular Biology, University of British Columbia, BC V6T 1Z4, Vancouver, Canada.
| | - Michael J Hogan
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Solna, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Norbert Pardi
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
34
|
Kinsey C, Lu T, Deiss A, Vuolo K, Klein L, Rustandi RR, Loughney JW. Determination of lipid content and stability in lipid nanoparticles using ultra high-performance liquid chromatography in combination with a Corona Charged Aerosol Detector. Electrophoresis 2022; 43:1091-1100. [PMID: 34784061 PMCID: PMC8652870 DOI: 10.1002/elps.202100244] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022]
Abstract
For many years, lipid nanoparticles (LNPs) have been used as delivery vehicles for various payloads (especially various oligonucleotides and mRNA), finding numerous applications in drug and vaccine development. LNP stability and bilayer fluidity are determined by the identities and the amounts of the various lipids employed in the formulation and LNP efficacy is determined in large part by the lipid composition which usually contains a cationic lipid, a PEG-lipid conjugate, cholesterol, and a zwitterionic helper phospholipid. Analytical methods developed for LNP characterization must be able to determine not only the identity and content of each individual lipid component (i.e., the parent lipids), but also the associated impurities and degradants. In this work, we describe an efficient and sensitive reversed-phase chromatographic method with charged aerosol detection (CAD) suitable for this purpose. Sample preparation diluent and mobile phase pH conditions are critical and have been optimized for the lipids of interest. This method was validated for its linearity, accuracy, precision, and specificity for lipid analysis to support process and formulation development for new drugs and vaccines.
Collapse
Affiliation(s)
- Caleb Kinsey
- Vaccine Analytical Research & DevelopmentMerck & Co. Inc.West PointPennsylvaniaUSA
| | - Tian Lu
- Vaccine Analytical Research & DevelopmentMerck & Co. Inc.West PointPennsylvaniaUSA
| | - Alyssa Deiss
- Vaccine Analytical Research & DevelopmentMerck & Co. Inc.West PointPennsylvaniaUSA
| | - Kim Vuolo
- Vaccine Analytical Research & DevelopmentMerck & Co. Inc.West PointPennsylvaniaUSA
| | - Lee Klein
- Vaccine Analytical Research & DevelopmentMerck & Co. Inc.West PointPennsylvaniaUSA
| | - Richard R. Rustandi
- Vaccine Analytical Research & DevelopmentMerck & Co. Inc.West PointPennsylvaniaUSA
| | - John W. Loughney
- Vaccine Analytical Research & DevelopmentMerck & Co. Inc.West PointPennsylvaniaUSA
| |
Collapse
|
35
|
Ionizable Lipid Nanoparticles Enhanced the Synergistic Adjuvant Effect of CpG ODNs and QS21 in a Varicella Zoster Virus Glycoprotein E Subunit Vaccine. Pharmaceutics 2022; 14:pharmaceutics14050973. [PMID: 35631559 PMCID: PMC9143440 DOI: 10.3390/pharmaceutics14050973] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
Varicella zoster virus (VZV) causes two diseases: varicella upon primary infection and herpes zoster when latent viruses in the sensory ganglia reactivate. While varicella vaccines depend on humoral immunity to prevent VZV infection, cell-mediated immunity (CMI), which plays a therapeutic role in the control or elimination of reactivated VZV in infected cells, is decisive for zoster vaccine efficacy. As one of the most abundant glycoproteins of VZV, conserved glycoprotein E (gE) is essential for viral replication and transmission between ganglion cells, thus making it an ideal target subunit vaccine antigen; gE has been successfully used in the herpes zoster vaccine ShingrixTM on the market. In this report, we found that ionizable lipid nanoparticles (LNPs) approved by the Food and Drug Administration (FDA) as vectors for coronavirus disease 2019 (COVID-19) mRNA vaccines could enhance the synergistic adjuvant effect of CpG oligodeoxynucleotides (CpG ODNs) and QS21 on VZV-gE, affecting both humoral immunity and CMI. Vaccines made with these LNPs showed promise as varicella vaccines without a potential risk of herpes zoster, which identifies them as a novel type of herpes zoster vaccine similar to ShingrixTM. All of the components in this LNP-CpG-QS21 adjuvant system were proven to be safe after mass vaccination, and the high proportion of cholesterol contained in the LNPs was helpful for limiting the cytotoxicity induced by QS21, which may lead to the development of a novel herpes zoster subunit vaccine for clinical application.
Collapse
|
36
|
Mitarotonda R, Giorgi E, Eufrasio-da-Silva T, Dolatshahi-Pirouz A, Mishra YK, Khademhosseini A, Desimone MF, De Marzi M, Orive G. Immunotherapeutic nanoparticles: From autoimmune disease control to the development of vaccines. BIOMATERIALS ADVANCES 2022; 135:212726. [PMID: 35475005 PMCID: PMC9023085 DOI: 10.1016/j.bioadv.2022.212726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/09/2022] [Accepted: 02/18/2022] [Indexed: 11/01/2022]
Abstract
The development of nanoparticles (NPs) with potential therapeutic uses represents an area of vast interest in the scientific community during the last years. Recently, the pandemic caused by COVID-19 motivated a race for vaccines creation to overcome the crisis generated. This is a good demonstration that nanotechnology will most likely be the basis of future immunotherapy. Moreover, the number of publications based on nanosystems has significantly increased in recent years and it is expected that most of these developments can go on to experimentation in clinical stages soon. The therapeutic use of NPs to combat different diseases such as cancer, allergies or autoimmune diseases will depend on their characteristics, their targets, and the transported molecules. This review presents an in-depth analysis of recent advances that have been developed in order to obtain novel nanoparticulate based tools for the treatment of allergies, autoimmune diseases and for their use in vaccines. Moreover, it is highlighted that by providing targeted delivery an increase in the potential of vaccines to induce an immune response is expected in the future. Definitively, the here gathered analysis is a good demonstration that nanotechnology will be the basis of future immunotherapy.
Collapse
Affiliation(s)
- Romina Mitarotonda
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina
| | - Exequiel Giorgi
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina
| | - Tatiane Eufrasio-da-Silva
- Department of Health Technology, Technical University of Denmark (DTU), 2800 Kgs. Lyngby, Denmark; Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Regenerative Biomaterials, Philips van Leydenlaan 25, 6525EX Nijmegen, the Netherlands
| | | | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, 6400 Sønderborg, Denmark
| | - Ali Khademhosseini
- Department of Bioengineering, Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Jonsson Comprehensive Cancer Center, Department of Radiology, University of California, Los Angeles, CA 90095, USA
| | - Martin F Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina.
| | - Mauricio De Marzi
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina.
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| |
Collapse
|
37
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), emerged in China in December 2019 and quickly spread around the globe, killing more than 4 million people and causing a severe economic crisis. This extraordinary situation prompted entities in government, industry, and academia to work together at unprecedented speed to develop safe and effective vaccines. Indeed, vaccines of multiple types have been generated in record time, and many have been evaluated in clinical trials. Of these, messenger RNA (mRNA) vaccines have emerged as lead candidates due to their speed of development and high degree of safety and efficacy. To date, two mRNA vaccines have received approval for human use, providing proof of the feasibility of this next-generation vaccine modality. This review gives a detailed overview about the types of mRNA vaccines developed for SARS-CoV-2, discusses and compares preclinical and clinical data, gives a mechanistic overview about immune responses generated by mRNA vaccination, and speculates on the challenges and promising future of this emergent vaccine platform.
Collapse
Affiliation(s)
- Michael J Hogan
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;
| | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
38
|
Zhang L, Liang Z, Chen C, Yang X, Fu D, Bao H, Li M, Shi S, Yu G, Zhang Y, Zhang C, Zhang W, Xue C, Sun B. Engineered Hydroxyapatite Nanoadjuvants with Controlled Shape and Aspect Ratios Reveal Their Immunomodulatory Potentials. ACS APPLIED MATERIALS & INTERFACES 2021; 13:59662-59672. [PMID: 34894655 DOI: 10.1021/acsami.1c17804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hydroxyapatite (HAP) has been formulated as adjuvants in vaccines for human use. However, the optimal properties required for HAP nanoparticles to elicit adjuvanticity and the underlying immunopotentiation mechanisms have not been fully elucidated. Herein, a library of HAP nanorods and nanospheres was synthesized to explore the effect of the particle shape and aspect ratio on the immune responses in vitro and adjuvanticity in vivo. It was demonstrated that long aspect ratio HAP nanorods induced a higher degree of cell membrane depolarization and subsequent uptake, and the internalized particles elicited cathepsin B release and mitochondrial reactive oxygen species generation, which further led to pro-inflammatory responses. Furthermore, the physicochemical property-dependent immunostimulation capacities were correlated with their humoral responses in a murine hepatitis B surface antigen immunization model, with long aspect ratio HAP nanorods inducing higher antigen-specific antibody productions. Importantly, HAP nanorods significantly up-regulated the IFN-γ secretion and CD107α expression on CD8+ T cells in immunized mice. Further mechanistic studies demonstrated that HAP nanorods with defined properties exerted immunomodulatory effects by enhanced antigen persistence and immune cell recruitments. Our study provides a rational design strategy for engineered nanomaterial-based vaccine adjuvants.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Zhihui Liang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Chen Chen
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Xuecheng Yang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Duo Fu
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Hang Bao
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Min Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Shuting Shi
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Ge Yu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Yixuan Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Caiqiao Zhang
- NCPC Genetech Biotechnology Co., Ltd., Shijiazhuang 050035, P. R. China
| | - Weiting Zhang
- NCPC Genetech Biotechnology Co., Ltd., Shijiazhuang 050035, P. R. China
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| |
Collapse
|
39
|
Li H, Monslow MA, Freed DC, Chang D, Li F, Gindy M, Wang D, Vora K, Espeseth AS, Petrovsky N, Fu TM. Novel adjuvants enhance immune responses elicited by a replication-defective human cytomegalovirus vaccine in nonhuman primates. Vaccine 2021; 39:7446-7456. [PMID: 34852943 DOI: 10.1016/j.vaccine.2021.10.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/13/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Adjuvants have long been explored to enhance vaccine efficacy. Current adjuvants approved for human vaccines are mostly studied for their ability to improve antibody responses. There remains a need for development of novel adjuvants, especially those able to enhance cell-mediated immunity (CMI). In this preclinical study we assessed the effect of two novel adjuvants, a delta inulin microparticle Advax formulated with or without a toll-like receptor 9 (TLR9) agonist CpG oligonucleotide, and a Merck & Co., Inc., Kenilworth, NJ, USA proprietary lipid nanoparticle (LNP), on immune responses elicited by V160, an experimental replication-defective human cytomegalovirus vaccine. Adult rhesus macaques were immunized with a low dose of V160 (10 units) either alone or in combination with the adjuvants as compared to those immunized with a high dose of V160 alone (100 units). While neither adjuvant conferred a significant benefit to vaccine-elicited humoral immune responses at the dose tested, both enhanced cellular immune responses to V160, where Advax promoted both CD4+ and CD8+ T cells and LNP predominantly impacted the CD4+ T cell response. Transcriptome analyses of peripheral blood samples demonstrated different modes of action for these adjuvants. One day post vaccination, LNP induced upregulation of a large number of genes involved in the innate immune response similar to those triggered by viral infection. In contrast, Advax did not activate any known inflammatory pathways and did not significantly impact gene expression pattern until day 7 post administration, suggesting a unique, non-inflammatory mechanism. These data warrant further exploration of Advax and LNP as adjuvants in clinical trials for vaccines desiring to elicit both humoral and T cell responses.
Collapse
Affiliation(s)
- Hualin Li
- Merck & Co., Inc., Kenilworth, NJ, USA.
| | | | | | - Dan Chang
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Dai Wang
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Nikolai Petrovsky
- Vaxine Pty Ltd, Flinders University, Bedford Park SA 5042, Australia
| | | |
Collapse
|
40
|
Alameh MG, Tombácz I, Bettini E, Lederer K, Sittplangkoon C, Wilmore JR, Gaudette BT, Soliman OY, Pine M, Hicks P, Manzoni TB, Knox JJ, Johnson JL, Laczkó D, Muramatsu H, Davis B, Meng W, Rosenfeld AM, Strohmeier S, Lin PJC, Mui BL, Tam YK, Karikó K, Jacquet A, Krammer F, Bates P, Cancro MP, Weissman D, Luning Prak ET, Allman D, Locci M, Pardi N. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity 2021; 54:2877-2892.e7. [PMID: 34852217 PMCID: PMC8566475 DOI: 10.1016/j.immuni.2021.11.001] [Citation(s) in RCA: 399] [Impact Index Per Article: 99.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 07/29/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022]
Abstract
Adjuvants are critical for improving the quality and magnitude of adaptive immune responses to vaccination. Lipid nanoparticle (LNP)-encapsulated nucleoside-modified mRNA vaccines have shown great efficacy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but the mechanism of action of this vaccine platform is not well-characterized. Using influenza virus and SARS-CoV-2 mRNA and protein subunit vaccines, we demonstrated that our LNP formulation has intrinsic adjuvant activity that promotes induction of strong T follicular helper cell, germinal center B cell, long-lived plasma cell, and memory B cell responses that are associated with durable and protective antibodies in mice. Comparative experiments demonstrated that this LNP formulation outperformed a widely used MF59-like adjuvant, AddaVax. The adjuvant activity of the LNP relies on the ionizable lipid component and on IL-6 cytokine induction but not on MyD88- or MAVS-dependent sensing of LNPs. Our study identified LNPs as a versatile adjuvant that enhances the efficacy of traditional and next-generation vaccine platforms.
Collapse
Affiliation(s)
| | - István Tombácz
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Bettini
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katlyn Lederer
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chutamath Sittplangkoon
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Joel R Wilmore
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian T Gaudette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ousamah Y Soliman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Pine
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip Hicks
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tomaz B Manzoni
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James J Knox
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John L Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dorottya Laczkó
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hiromi Muramatsu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Davis
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenzhao Meng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aaron M Rosenfeld
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Katalin Karikó
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA; BioNTech RNA Pharmaceuticals, Mainz, Germany
| | - Alain Jacquet
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Cancro
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michela Locci
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Antonarelli G, Corti C, Tarantino P, Ascione L, Cortes J, Romero P, Mittendorf EA, Disis ML, Curigliano G. Therapeutic cancer vaccines revamping: technology advancements and pitfalls. Ann Oncol 2021; 32:1537-1551. [PMID: 34500046 PMCID: PMC8420263 DOI: 10.1016/j.annonc.2021.08.2153] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/21/2021] [Accepted: 08/29/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer vaccines (CVs) represent a long-sought therapeutic and prophylactic immunotherapy strategy to obtain antigen (Ag)-specific T-cell responses and potentially achieve long-term clinical benefit. However, historically, most CV clinical trials have resulted in disappointing outcomes, despite promising signs of immunogenicity across most formulations. In the past decade, technological advances regarding vaccine delivery platforms, tools for immunogenomic profiling, and Ag/epitope selection have occurred. Consequently, the ability of CVs to induce tumor-specific and, in some cases, remarkable clinical responses have been observed in early-phase clinical trials. It is notable that the record-breaking speed of vaccine development in response to the coronavirus disease-2019 pandemic mainly relied on manufacturing infrastructures and technological platforms already developed for CVs. In turn, research, clinical data, and infrastructures put in place for the severe acute respiratory syndrome coronavirus 2 pandemic can further speed CV development processes. This review outlines the main technological advancements as well as major issues to tackle in the development of CVs. Possible applications for unmet clinical needs will be described, putting into perspective the future of cancer vaccinology.
Collapse
Affiliation(s)
- G Antonarelli
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - C Corti
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - P Tarantino
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - L Ascione
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - J Cortes
- International Breast Cancer Center (IBCC), Quironsalud Group, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - P Romero
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
| | - E A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, USA; Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, USA
| | - M L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, USA
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
42
|
Swaminathan G, Citron M, Xiao J, Norton JE, Reens AL, Topçuoğlu BD, Maritz JM, Lee KJ, Freed DC, Weber TM, White CH, Kadam M, Spofford E, Bryant-Hall E, Salituro G, Kommineni S, Liang X, Danilchanka O, Fontenot JA, Woelk CH, Gutierrez DA, Hazuda DJ, Hannigan GD. Vaccine Hyporesponse Induced by Individual Antibiotic Treatment in Mice and Non-Human Primates Is Diminished upon Recovery of the Gut Microbiome. Vaccines (Basel) 2021; 9:vaccines9111340. [PMID: 34835271 PMCID: PMC8619314 DOI: 10.3390/vaccines9111340] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
Emerging evidence demonstrates a connection between microbiome composition and suboptimal response to vaccines (vaccine hyporesponse). Harnessing the interaction between microbes and the immune system could provide novel therapeutic strategies for improving vaccine response. Currently we do not fully understand the mechanisms and dynamics by which the microbiome influences vaccine response. Using both mouse and non-human primate models, we report that short-term oral treatment with a single antibiotic (vancomycin) results in the disruption of the gut microbiome and this correlates with a decrease in systemic levels of antigen-specific IgG upon subsequent parenteral vaccination. We further show that recovery of microbial diversity before vaccination prevents antibiotic-induced vaccine hyporesponse, and that the antigen specific IgG response correlates with the recovery of microbiome diversity. RNA sequencing analysis of small intestine, spleen, whole blood, and secondary lymphoid organs from antibiotic treated mice revealed a dramatic impact on the immune system, and a muted inflammatory signature is correlated with loss of bacteria from Lachnospiraceae, Ruminococcaceae, and Clostridiaceae. These results suggest that microbially modulated immune pathways may be leveraged to promote vaccine response and will inform future vaccine design and development strategies.
Collapse
Affiliation(s)
- Gokul Swaminathan
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
- Correspondence: (G.S.); (G.D.H.)
| | - Michael Citron
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Jianying Xiao
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - James E. Norton
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Abigail L. Reens
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Begüm D. Topçuoğlu
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Julia M. Maritz
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Keun-Joong Lee
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, MRL, Merck & Co. Inc., Rahway, NJ 07065, USA; (K.-J.L.); (G.S.)
| | - Daniel C. Freed
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Teresa M. Weber
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Cory H. White
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Mahika Kadam
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Erin Spofford
- Safety Assessment and Laboratory Animal Research, MRL, Merck & Co. Inc., Boston, MA 02115, USA; (E.S.); (E.B.-H.)
| | - Erin Bryant-Hall
- Safety Assessment and Laboratory Animal Research, MRL, Merck & Co. Inc., Boston, MA 02115, USA; (E.S.); (E.B.-H.)
| | - Gino Salituro
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, MRL, Merck & Co. Inc., Rahway, NJ 07065, USA; (K.-J.L.); (G.S.)
| | - Sushma Kommineni
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Olga Danilchanka
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Jane A. Fontenot
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, LA 70503, USA;
| | - Christopher H. Woelk
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Dario A. Gutierrez
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
| | - Daria J. Hazuda
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
- Infectious Diseases and Vaccine Research, MRL, Merck & Co., Inc., West Point, PA 19486, USA; (M.C.); (J.X.); (D.C.F.); (T.M.W.)
| | - Geoffrey D. Hannigan
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA; (J.E.N.J.); (A.L.R.); (B.D.T.); (J.M.M.); (C.H.W.); (M.K.); (S.K.); (X.L.); (O.D.); (C.H.W.); (D.A.G.); (D.J.H.)
- Correspondence: (G.S.); (G.D.H.)
| |
Collapse
|
43
|
Stater EP, Sonay AY, Hart C, Grimm J. The ancillary effects of nanoparticles and their implications for nanomedicine. NATURE NANOTECHNOLOGY 2021; 16:1180-1194. [PMID: 34759355 PMCID: PMC9031277 DOI: 10.1038/s41565-021-01017-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/22/2021] [Indexed: 05/12/2023]
Abstract
Nanoparticles are often engineered as a scaffolding system to combine targeting, imaging and/or therapeutic moieties into a unitary agent. However, mostly overlooked, the nanomaterial itself interacts with biological systems exclusive of application-specific particle functionalization. This nanoparticle biointerface has been found to elicit specific biological effects, which we term 'ancillary effects'. In this Review, we describe the current state of knowledge of nanobiology gleaned from existing studies of ancillary effects with the objectives to describe the potential of nanoparticles to modulate biological effects independently of any engineered function; evaluate how these effects might be relevant for nanomedicine design and functional considerations, particularly how they might be useful to inform clinical decision-making; identify potential clinical harm that arises from adverse nanoparticle interactions with biology; and, finally, highlight the current lack of knowledge in this area as both a barrier and an incentive to the further development of nanomedicine.
Collapse
Affiliation(s)
- Evan P Stater
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Ali Y Sonay
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cassidy Hart
- Department of General Surgery, Lankenau Medical Center, Wynnewood, PA, USA
| | - Jan Grimm
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
44
|
Pilkington EH, Suys EJA, Trevaskis NL, Wheatley AK, Zukancic D, Algarni A, Al-Wassiti H, Davis TP, Pouton CW, Kent SJ, Truong NP. From influenza to COVID-19: Lipid nanoparticle mRNA vaccines at the frontiers of infectious diseases. Acta Biomater 2021; 131:16-40. [PMID: 34153512 PMCID: PMC8272596 DOI: 10.1016/j.actbio.2021.06.023] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023]
Abstract
Vaccination represents the best line of defense against infectious diseases and is crucial in curtailing pandemic spread of emerging pathogens to which a population has limited immunity. In recent years, mRNA vaccines have been proposed as the new frontier in vaccination, owing to their facile and rapid development while providing a safer alternative to traditional vaccine technologies such as live or attenuated viruses. Recent breakthroughs in mRNA vaccination have been through formulation with lipid nanoparticles (LNPs), which provide both protection and enhanced delivery of mRNA vaccines in vivo. In this review, current paradigms and state-of-the-art in mRNA-LNP vaccine development are explored through first highlighting advantages posed by mRNA vaccines, establishing LNPs as a biocompatible delivery system, and finally exploring the use of mRNA-LNP vaccines in vivo against infectious disease towards translation to the clinic. Furthermore, we highlight the progress of mRNA-LNP vaccine candidates against COVID-19 currently in clinical trials, with the current status and approval timelines, before discussing their future outlook and challenges that need to be overcome towards establishing mRNA-LNPs as next-generation vaccines. STATEMENT OF SIGNIFICANCE: With the recent success of mRNA vaccines developed by Moderna and BioNTech/Pfizer against COVID-19, mRNA technology and lipid nanoparticles (LNP) have never received more attention. This manuscript timely reviews the most advanced mRNA-LNP vaccines that have just been approved for emergency use and are in clinical trials, with a focus on the remarkable development of several COVID-19 vaccines, faster than any other vaccine in history. We aim to give a comprehensive introduction of mRNA and LNP technology to the field of biomaterials science and increase accessibility to readers with a new interest in mRNA-LNP vaccines. We also highlight current limitations and future outlook of the mRNA vaccine technology that need further efforts of biomaterials scientists to address.
Collapse
Affiliation(s)
- Emily H Pilkington
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Estelle J A Suys
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Natalie L Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Danijela Zukancic
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Azizah Algarni
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Hareth Al-Wassiti
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Australia
| | - Colin W Pouton
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Nghia P Truong
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3000, Australia.
| |
Collapse
|
45
|
Han S, Mei L, Quach T, Porter C, Trevaskis N. Lipophilic Conjugates of Drugs: A Tool to Improve Drug Pharmacokinetic and Therapeutic Profiles. Pharm Res 2021; 38:1497-1518. [PMID: 34463935 DOI: 10.1007/s11095-021-03093-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/05/2021] [Indexed: 01/19/2023]
Abstract
Lipophilic conjugates (LCs) of small molecule drugs have been used widely in clinical and pre-clinical studies to achieve a number of pharmacokinetic and therapeutic benefits. For example, lipophilic derivatives of drugs are employed in several long acting injectable products to provide sustained drug exposure for hormone replacement therapy and to treat conditions such as neuropsychiatric diseases. LCs can also be used to modulate drug metabolism, and to enhance drug permeation across membranes, either by increasing lipophilicity to enhance passive diffusion or by increasing protein-mediated active transport. Furthermore, such conjugation strategies have been employed to promote drug association with endogenous macromolecular carriers (e.g. albumin and lipoproteins), and this in turn results in altered drug distribution and pharmacokinetic profiles, where the changes can be 'general' (e.g. prolonged plasma half-life) or 'specific' (e.g. enhanced delivery to specific tissues in parallel with the macromolecular carriers). Another utility of LCs is to enhance the encapsulation of drugs within engineered nanoscale drug delivery systems, in order to best take advantage of the targeting and pharmacokinetic benefits of nanomedicines. The current review provides a summary of the mechanisms by which lipophilic conjugates, including in combination with delivery vehicles, can be used to control drug delivery, distribution and therapeutic profiles. The article is structured into sections which highlight a specific benefit of LCs and then demonstrate this benefit with case studies. The review attempts to provide a toolbox to assist researchers to design and optimise drug candidates, including consideration of drug-formulation compatibility.
Collapse
Affiliation(s)
- Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
- Suzhou Institute of Drug Innovation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China.
| | - Lianghe Mei
- Suzhou Institute of Drug Innovation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Tim Quach
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- PureTech Health, 6 Tide Street, Boston, MA, 02210, USA
| | - Chris Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Natalie Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
| |
Collapse
|
46
|
Chatzikleanthous D, O'Hagan DT, Adamo R. Lipid-Based Nanoparticles for Delivery of Vaccine Adjuvants and Antigens: Toward Multicomponent Vaccines. Mol Pharm 2021; 18:2867-2888. [PMID: 34264684 DOI: 10.1021/acs.molpharmaceut.1c00447] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite the many advances that have occurred in the field of vaccine adjuvants, there are still unmet needs that may enable the development of vaccines suitable for more challenging pathogens (e.g., HIV and tuberculosis) and for cancer vaccines. Liposomes have already been shown to be highly effective as adjuvant/delivery systems due to their versatility and likely will find further uses in this space. The broad potential of lipid-based delivery systems is highlighted by the recent approval of COVID-19 vaccines comprising lipid nanoparticles with encapsulated mRNA. This review provides an overview of the different approaches that can be evaluated for the design of lipid-based vaccine adjuvant/delivery systems for protein, carbohydrate, and nucleic acid-based antigens and how these strategies might be combined to develop multicomponent vaccines.
Collapse
Affiliation(s)
- Despo Chatzikleanthous
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, G4 0RE Glasgow, U.K.,GSK, Via Fiorentina 1, 53100 Siena, Italy
| | | | | |
Collapse
|
47
|
Igyártó BZ, Jacobsen S, Ndeupen S. Future considerations for the mRNA-lipid nanoparticle vaccine platform. Curr Opin Virol 2021; 48:65-72. [PMID: 33906124 PMCID: PMC8065267 DOI: 10.1016/j.coviro.2021.03.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 12/16/2022]
Abstract
Vaccines based on mRNA-containing lipid nanoparticles (LNPs) pioneered by Katalin Karikó and Drew Weissman at the University of Pennsylvania are a promising new vaccine platform used by two of the leading vaccines against coronavirus disease in 2019 (COVID-19). However, there are many questions regarding their mechanism of action in humans that remain unanswered. Here we consider the immunological features of LNP components and off-target effects of the mRNA, both of which could increase the risk of side effects. We suggest ways to mitigate these potential risks by harnessing dendritic cell (DC) biology.
Collapse
Affiliation(s)
- Botond Z Igyártó
- Thomas Jefferson University, Department of Microbiology and Immunology, 233 South 10th Street, Philadelphia, PA 19107, United States.
| | - Sonya Jacobsen
- Thomas Jefferson University, Department of Microbiology and Immunology, 233 South 10th Street, Philadelphia, PA 19107, United States
| | - Sonia Ndeupen
- Thomas Jefferson University, Department of Microbiology and Immunology, 233 South 10th Street, Philadelphia, PA 19107, United States
| |
Collapse
|
48
|
Kowalzik F, Schreiner D, Jensen C, Teschner D, Gehring S, Zepp F. mRNA-Based Vaccines. Vaccines (Basel) 2021; 9:390. [PMID: 33921028 PMCID: PMC8103517 DOI: 10.3390/vaccines9040390] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022] Open
Abstract
Increases in the world's population and population density promote the spread of emerging pathogens. Vaccines are the most cost-effective means of preventing this spread. Traditional methods used to identify and produce new vaccines are not adequate, in most instances, to ensure global protection. New technologies are urgently needed to expedite large scale vaccine development. mRNA-based vaccines promise to meet this need. mRNA-based vaccines exhibit a number of potential advantages relative to conventional vaccines, namely they (1) involve neither infectious elements nor a risk of stable integration into the host cell genome; (2) generate humoral and cell-mediated immunity; (3) are well-tolerated by healthy individuals; and (4) are less expensive and produced more rapidly by processes that are readily standardized and scaled-up, improving responsiveness to large emerging outbreaks. Multiple mRNA vaccine platforms have demonstrated efficacy in preventing infectious diseases and treating several types of cancers in humans as well as animal models. This review describes the factors that contribute to maximizing the production of effective mRNA vaccine transcripts and delivery systems, and the clinical applications are discussed in detail.
Collapse
Affiliation(s)
- Frank Kowalzik
- Pediatric Department, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany; (D.S.); (C.J.); (S.G.); (F.Z.)
| | - Daniel Schreiner
- Pediatric Department, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany; (D.S.); (C.J.); (S.G.); (F.Z.)
| | - Christian Jensen
- Pediatric Department, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany; (D.S.); (C.J.); (S.G.); (F.Z.)
| | - Daniel Teschner
- Department of Hematology, Medical Oncology, and Pneumology, University Medical Center of the Johannes Gutenberg University, 55122 Mainz, Germany;
| | - Stephan Gehring
- Pediatric Department, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany; (D.S.); (C.J.); (S.G.); (F.Z.)
| | - Fred Zepp
- Pediatric Department, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany; (D.S.); (C.J.); (S.G.); (F.Z.)
| |
Collapse
|
49
|
Thi TTH, Suys EJA, Lee JS, Nguyen DH, Park KD, Truong NP. Lipid-Based Nanoparticles in the Clinic and Clinical Trials: From Cancer Nanomedicine to COVID-19 Vaccines. Vaccines (Basel) 2021; 9:359. [PMID: 33918072 PMCID: PMC8069344 DOI: 10.3390/vaccines9040359] [Citation(s) in RCA: 273] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
COVID-19 vaccines have been developed with unprecedented speed which would not have been possible without decades of fundamental research on delivery nanotechnology. Lipid-based nanoparticles have played a pivotal role in the successes of COVID-19 vaccines and many other nanomedicines, such as Doxil® and Onpattro®, and have therefore been considered as the frontrunner in nanoscale drug delivery systems. In this review, we aim to highlight the progress in the development of these lipid nanoparticles for various applications, ranging from cancer nanomedicines to COVID-19 vaccines. The lipid-based nanoparticles discussed in this review are liposomes, niosomes, transfersomes, solid lipid nanoparticles, and nanostructured lipid carriers. We particularly focus on the innovations that have obtained regulatory approval or that are in clinical trials. We also discuss the physicochemical properties required for specific applications, highlight the differences in requirements for the delivery of different cargos, and introduce current challenges that need further development. This review serves as a useful guideline for designing new lipid nanoparticles for both preventative and therapeutic vaccines including immunotherapies.
Collapse
Affiliation(s)
- Thai Thanh Hoang Thi
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| | - Estelle J. A. Suys
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia;
| | - Jung Seok Lee
- Biomedical Engineering, Malone Engineering Center 402A, Yale University, 55 Prospect St., New Haven, CT 06511, USA;
| | - Dai Hai Nguyen
- Vietnam Academy of Science and Technology, Graduate University of Science and Technology, Hanoi 100000, Vietnam;
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 01 TL29 District 12, Ho Chi Minh City 700000, Vietnam
| | - Ki Dong Park
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea;
| | - Nghia P. Truong
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia;
| |
Collapse
|
50
|
Wollner CJ, Richner JM. mRNA Vaccines against Flaviviruses. Vaccines (Basel) 2021; 9:148. [PMID: 33673131 PMCID: PMC7918459 DOI: 10.3390/vaccines9020148] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Numerous vaccines have now been developed using the mRNA platform. In this approach, mRNA coding for a viral antigen is in vitro synthesized and injected into the host leading to exogenous protein expression and robust immune responses. Vaccines can be rapidly developed utilizing the mRNA platform in the face of emerging pandemics. Additionally, the mRNA coding region can be easily manipulated to test novel hypotheses in order to combat viral infections which have remained refractory to traditional vaccine approaches. Flaviviruses are a diverse family of viruses that cause widespread disease and have pandemic potential. In this review, we discuss the mRNA vaccines which have been developed against diverse flaviviruses.
Collapse
Affiliation(s)
| | - Justin M. Richner
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|