Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1381508, 2024.
Article in English | MEDLINE | ID: mdl-38690272

ABSTRACT

Seasonal influenza remains a serious global health problem, leading to high mortality rates among the elderly and individuals with comorbidities. Vaccination is generally accepted as the most effective strategy for influenza prevention. While current influenza vaccines are effective, they still have limitations, including narrow specificity for certain serological variants, which may result in a mismatch between vaccine antigens and circulating strains. Additionally, the rapid variability of the virus poses challenges in providing extended protection beyond a single season. Therefore, mRNA technology is particularly promising for influenza prevention, as it enables the rapid development of multivalent vaccines and allows for quick updates of their antigenic composition. mRNA vaccines have already proven successful in preventing COVID-19 by eliciting rapid cellular and humoral immune responses. In this study, we present the development of a trivalent mRNA vaccine candidate, evaluate its immunogenicity using the hemagglutination inhibition assay, ELISA, and assess its efficacy in animals. We demonstrate the higher immunogenicity of the mRNA vaccine candidate compared to the inactivated split influenza vaccine and its enhanced ability to generate a cross-specific humoral immune response. These findings highlight the potential mRNA technology in overcoming current limitations of influenza vaccines and hold promise for ensuring greater efficacy in preventing seasonal influenza outbreaks.


Subject(s)
Immunity, Humoral , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , mRNA Vaccines , Animals , Female , Humans , Mice , Cross Reactions/immunology , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Humoral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice, Inbred BALB C , mRNA Vaccines/administration & dosage , mRNA Vaccines/chemistry , mRNA Vaccines/genetics , mRNA Vaccines/immunology , Seasons , Time Factors , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
2.
Adv Mater ; 36(23): e2311574, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38433564

ABSTRACT

Dendritic cell (DC) maturation is a crucial process for antigen presentation and the initiation of T cell-mediated immune responses. Toll-like receptors play pivotal roles in stimulating DC maturation and promoting antigen presentation. Here, a novel message RNA (mRNA) cancer vaccine is reported that boosts antitumor efficacy by codelivering an mRNA encoding tumor antigen and a TLR7/8 agonist (R848) to DC using supramolecular lipid nanoparticles (SMLNP) as a delivery platform, in which a new ionizable lipid (N2-3L) remarkably enhances the translation efficiency of mRNA and a ß-cyclodextrin (ß-CD)-modified ionizable lipid (Lip-CD) encapsulates R848. The incorporation of R848 adjuvant into the mRNA vaccine through noncovalent host-guest complexation significantly promotes DC maturation and antigen presentation after vaccination, thus resulting in superior antitumor efficacy in vivo. Moreover, the antitumor efficacy is further boosted synergized with immune checkpoint blockade by potentiating the anticancer capability of cytotoxic T lymphocytes infiltrated in tumor sites. This work indicates that SMLNP shows brilliant potential as next-generation delivery system in the development of mRNA vaccines with high efficacy.


Subject(s)
Cancer Vaccines , Dendritic Cells , Imidazoles , Immunotherapy , Lipids , Nanoparticles , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Animals , Nanoparticles/chemistry , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Dendritic Cells/immunology , Mice , Lipids/chemistry , Imidazoles/chemistry , mRNA Vaccines/chemistry , beta-Cyclodextrins/chemistry , RNA, Messenger/genetics , RNA, Messenger/chemistry , Neoplasms/therapy , Cell Line, Tumor , Antigens, Neoplasm/immunology , Humans , Mice, Inbred C57BL , Liposomes
3.
Nature ; 621(7978): 396-403, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37130545

ABSTRACT

Messenger RNA (mRNA) vaccines are being used to combat the spread of COVID-19 (refs. 1-3), but they still exhibit critical limitations caused by mRNA instability and degradation, which are major obstacles for the storage, distribution and efficacy of the vaccine products4. Increasing secondary structure lengthens mRNA half-life, which, together with optimal codons, improves protein expression5. Therefore, a principled mRNA design algorithm must optimize both structural stability and codon usage. However, owing to synonymous codons, the mRNA design space is prohibitively large-for example, there are around 2.4 × 10632 candidate mRNA sequences for the SARS-CoV-2 spike protein. This poses insurmountable computational challenges. Here we provide a simple and unexpected solution using the classical concept of lattice parsing in computational linguistics, where finding the optimal mRNA sequence is analogous to identifying the most likely sentence among similar-sounding alternatives6. Our algorithm LinearDesign finds an optimal mRNA design for the spike protein in just 11 minutes, and can concurrently optimize stability and codon usage. LinearDesign substantially improves mRNA half-life and protein expression, and profoundly increases antibody titre by up to 128 times in mice compared to the codon-optimization benchmark on mRNA vaccines for COVID-19 and varicella-zoster virus. This result reveals the great potential of principled mRNA design and enables the exploration of previously unreachable but highly stable and efficient designs. Our work is a timely tool for vaccines and other mRNA-based medicines encoding therapeutic proteins such as monoclonal antibodies and anti-cancer drugs7,8.


Subject(s)
Algorithms , COVID-19 Vaccines , COVID-19 , RNA Stability , RNA, Messenger , SARS-CoV-2 , mRNA Vaccines , Animals , Humans , Mice , Codon/genetics , COVID-19/genetics , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/chemistry , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Half-Life , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/immunology , mRNA Vaccines/chemistry , mRNA Vaccines/genetics , mRNA Vaccines/immunology , RNA Stability/genetics , RNA Stability/immunology , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/immunology
5.
Science ; 376(6594): 680-681, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35549401

ABSTRACT

New delivery systems aim to increase vaccine potency and reduce side effects.


Subject(s)
Lipids , Nanoparticles , mRNA Vaccines , Humans , Lipids/chemistry , Nanoparticles/adverse effects , Nanoparticles/chemistry , mRNA Vaccines/administration & dosage , mRNA Vaccines/chemistry
7.
Chem Pharm Bull (Tokyo) ; 69(12): 1141-1159, 2021.
Article in English | MEDLINE | ID: mdl-34853281

ABSTRACT

Considerable efforts have been made on the development of lipid nanoparticles (LNPs) for delivering of nucleic acids in LNP-based medicines, including a first-ever short interfering RNA (siRNA) medicine, Onpattro, and the mRNA vaccines against the coronavirus disease 2019 (COVID-19), which have been approved and are currently in use worldwide. The successful rational design of ionizable cationic lipids was a major breakthrough that dramatically increased delivery efficiency in this field. The LNPs would be expected to be useful as a platform technology for the delivery of various therapeutic modalities for genome editing and even for undiscovered therapeutic mechanisms. In this review, the current progress of my research, including the molecular design of pH-sensitive cationic lipids, their applications for various tissues and cell types, and for delivering various macromolecules, including siRNA, antisense oligonucleotide, mRNA, and the clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) system will be described. Mechanistic studies regarding relationships between the physicochemical properties of LNPs, drug delivery, and biosafety are also summarized. Furthermore, current issues that need to be addressed for next generation drug delivery systems are discussed.


Subject(s)
Drug Carriers/chemistry , Lipids/chemistry , Liposomes/chemistry , Nanoparticles/chemistry , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Cations/chemistry , Hydrogen-Ion Concentration , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , SARS-CoV-2/isolation & purification , mRNA Vaccines/chemistry , mRNA Vaccines/metabolism
8.
Acc Chem Res ; 54(23): 4262-4271, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34756014

ABSTRACT

Over the past two decades, research on mRNA-based therapies has exploded, mainly because of the inherent advantages of mRNA, including a low integration probability, transient expression, and simple and rapid in vitro transcription production approaches. In addition, thanks to improved stability and reduced immunogenicity by advanced strategies, the application of mRNA has expanded from protein replacement therapy to vaccination, gene editing and other fields, showing great promise for clinical application. Recently, with the successive launch of two mRNA-based COVID-19 vaccines, mRNA technology has attracted an enormous amount of attention from scientific researchers as well as pharmaceutical companies. Because of the large molecular weight, hydrophilicity, and highly negative charge densities of mRNA, it is difficult to overcome the intracellular delivery barriers. Therefore, various delivery vehicles have been developed to achieve more effective mRNA delivery. In general, conventional mRNA administration methods are based on injection strategies, including intravenous, intramuscular, intradermal, and subcutaneous injections. Although these routes circumvent the absorption barriers to some extent, they bring about injection-related concerns such as safety issues, pain, low compliance, and difficulty in repeated dosing, increasing the need to explore alternative strategies for noninvasive delivery. The ideal noninvasive delivery systems are featured with easy to use, low risks of infection, and good patient compliance. At the same time, they allow patients to self-administer, reducing reliance on professional healthcare workers and interference with bodily functions and daily life. In particular, the noninvasive mucosal delivery of mRNA vaccines can induce mucosal immune responses, which are important for resisting pathogens infected through mucosal routes.Because of the potential clinical benefits mentioned above, we detailed the existing strategies for the noninvasive delivery of mRNA in this review, including delivery via the nasal, pulmonary, vaginal, and transdermal routes. First, we discussed the unique strengths and biological hindrances of each route on the basis of physiology. Next, we comprehensively summarized the research progress reported so far and analyzed the technologies and delivery vehicles used, hoping to provide some references for further explorations. Among these noninvasive routes, nasal and pulmonary delivery are the earliest and most intensively studied areas, mostly owing to their favorable physiological structures: the nasal or pulmonary mucosa is easily accessible, highly permeable and highly vascularized. In contrast, the development of vaginal mRNA delivery is relatively less reported, and the current research mainly focused on some local applications. In addition, microneedles have also been investigated to overcome skin barriers for mRNA delivery in recent years, making microneedle-based delivery an emerging alternative pathway. In summary, a variety of mRNA formulations and delivery strategies have been developed for noninvasive mRNA delivery, skillfully combining appropriate vehicles or physical technologies to enhance effectiveness. We surmise that continuous advances and technological innovations in the development of mRNA noninvasive delivery will accelerate the translation from experimental research to clinical application.


Subject(s)
Drug Delivery Systems/methods , mRNA Vaccines/chemistry , Administration, Cutaneous , Administration, Inhalation , Animals , COVID-19/prevention & control , COVID-19/virology , Humans , SARS-CoV-2/isolation & purification , mRNA Vaccines/administration & dosage , mRNA Vaccines/immunology
9.
Microbiol Spectr ; 9(3): e0096521, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34756082

ABSTRACT

The prompt rollout of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccine is facilitating population immunity, which is becoming more dominant than natural infection-mediated immunity. In the midst of coronavirus disease 2019 (COVID-19) vaccine deployment, understanding the epitope profiles of vaccine-elicited antibodies will be the first step in assessing the functionality of vaccine-induced immunity. In this study, the high-resolution linear epitope profiles of Pfizer-BioNTech COVID-19 mRNA vaccine recipients and COVID-19 patients were delineated by using microarrays mapped with overlapping peptides of the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. The vaccine-induced antibodies targeting the RBD had a broader distribution across the RBD than that induced by the natural infection. Half-maximal neutralization titers were measured in vitro by live virus neutralization assays. As a result, relatively lower neutralizability was observed in vaccine recipient sera, when normalized to a total anti-RBD IgG titer. However, mutation panel assays targeting the SARS-CoV-2 variants of concern have shown that the vaccine-induced epitope variety, rich in breadth, may grant resistance against future viral evolutionary escapes, serving as an advantage of vaccine-induced immunity. IMPORTANCE Establishing vaccine-based population immunity has been the key factor in attaining herd protection. Thanks to expedited worldwide research efforts, the potency of mRNA vaccines against the coronavirus disease 2019 (COVID-19) is now incontestable. The next debate is regarding the coverage of SARS-CoV-2 variants. In the midst of vaccine deployment, it is of importance to describe the similarities and differences between the immune responses of COVID-19 vaccine recipients and naturally infected individuals. In this study, we demonstrated that the antibody profiles of vaccine recipients are richer in variety, targeting a key protein of the invading virus, than those of naturally infected individuals. Vaccine-elicited antibodies included more nonneutralizing antibodies than infection-elicited antibodies, and their breadth in antibody variations suggested possible resilience against future SARS-CoV-2 variants. The antibody profile achieved by vaccinations in naive individuals provides important insight into the first step toward vaccine-based population immunity.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Epitope Mapping , Protein Binding , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/chemistry , mRNA Vaccines/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , COVID-19/prevention & control , COVID-19 Vaccines/chemistry , Humans , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Vaccines, Synthetic/immunology , mRNA Vaccines/chemistry
10.
Bioconjug Chem ; 32(12): 2497-2506, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34775749

ABSTRACT

Understanding immune responses toward viral infection will be useful for potential therapeutic intervention and offer insights into the design of prophylactic vaccines. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the COVID-19 pandemic. To understand the complex immune responses toward SARS-CoV-2 infection, here we developed a method to express and purify the recombinant and engineered viral receptor-binding domain (RBD) to more than 95% purity. We could encapsulate RNA molecules into the interior of a virion-sized liposome. We conjugated the purified RBD proteins onto the surface of the liposome in an orientation-specific manner with defined spatial densities. Both the encapsulation of RNAs and the chemical conjugation of the RBD protein on liposome surfaces were stable under physiologically relevant conditions. In contrast to soluble RBD proteins, a single injection of RBD-conjugated liposomes alone, in the absence of any other adjuvants, elicited RBD-specific B cell responses in BALB/c mice, and the resulting animal sera could potently neutralize HIV-1 pseudovirions that displayed the SARS-CoV-2 spike proteins. These results validate these supramolecular structures as a novel and effective tool to mimic the structure of enveloped viruses, the use of which will allow systematic dissection of the complex B cell responses to SARS-CoV-2 infection.


Subject(s)
Antibodies, Neutralizing/immunology , COVID-19 Vaccines/therapeutic use , COVID-19/prevention & control , Liposomes/therapeutic use , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/therapeutic use , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/therapeutic use , Animals , COVID-19/immunology , COVID-19 Vaccines/chemistry , Female , Humans , Immunization , Liposomes/chemistry , Mice, Inbred BALB C , Models, Molecular , Protein Domains , Spike Glycoprotein, Coronavirus/chemistry , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/therapeutic use , mRNA Vaccines/chemistry , mRNA Vaccines/therapeutic use
11.
J Am Chem Soc ; 143(49): 20529-20545, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34841867

ABSTRACT

Unquestionably, polymers have influenced the world over the past 100 years. They are now more crucial than ever since the COVID-19 pandemic outbreak. The pandemic paved the way for certain polymers to be in the spotlight, namely sequence-defined polymers such as messenger ribonucleic acid (mRNA), which was the first type of vaccine to be authorized in the U.S. and Europe to protect against the SARS-CoV-2 virus. This rise of mRNA will probably influence scientific research concerning nucleic acids in general and RNA therapeutics in specific. In this Perspective, we highlight the recent trends in sequence-controlled and sequence-defined polymers. Then we discuss mRNA vaccines as an example to illustrate the need of ultimate sequence control to achieve complex functions such as specific activation of the immune system. We briefly present how mRNA vaccines are produced, the importance of modified nucleotides, the characteristic features, and the advantages and challenges associated with this class of vaccines. Finally, we discuss the chances and opportunities for polymer chemistry to provide solutions and contribute to the future progress of RNA-based therapeutics. We highlight two particular roles of polymers in this context. One represents conjugation of polymers to nucleic acids to form biohybrids. The other is concerned with advanced polymer-based carrier systems for nucleic acids. We believe that polymers can help to address present problems of RNA-based therapeutic technologies and impact the field beyond the COVID-19 pandemic.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , COVID-19 Vaccines/chemistry , Polymers/pharmacology , SARS-CoV-2/drug effects , mRNA Vaccines/chemistry , Animals , Drug Carriers , Humans
12.
Nat Commun ; 12(1): 6777, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34811367

ABSTRACT

Lipid nanoparticle (LNP)-formulated mRNA vaccines were rapidly developed and deployed in response to the SARS-CoV-2 pandemic. Due to the labile nature of mRNA, identifying impurities that could affect product stability and efficacy is crucial to the long-term use of nucleic-acid based medicines. Herein, reversed-phase ion pair high performance liquid chromatography (RP-IP HPLC) was used to identify a class of impurity formed through lipid:mRNA reactions; such reactions are typically undetectable by traditional mRNA purity analytical techniques. The identified modifications render the mRNA untranslatable, leading to loss of protein expression. Specifically, electrophilic impurities derived from the ionizable cationic lipid component are shown to be responsible. Mechanisms implicated in the formation of reactive species include oxidation and subsequent hydrolysis of the tertiary amine. It thus remains critical to ensure robust analytical methods and stringent manufacturing control to ensure mRNA stability and high activity in LNP delivery systems.


Subject(s)
Drug Delivery Systems , Liposomes/chemistry , Nanoparticles/chemistry , RNA, Messenger/chemistry , Vaccine Potency , Aldehydes/chemistry , Chromatography, Liquid , Humans , Ions/chemistry , Lipids/chemistry , Nucleosides/chemistry , Oxidation-Reduction , Protein Biosynthesis , RNA Stability , mRNA Vaccines/chemistry
13.
Viruses ; 13(8)2021 07 27.
Article in English | MEDLINE | ID: mdl-34452327

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) infection affects millions of individuals worldwide and can lead to severe leukemia, myelopathy/tropical spastic paraparesis, and numerous other disorders. Pursuing a safe and effective immunotherapeutic approach, we compared the viral polyprotein and the human proteome with a sliding window approach in order to identify oligopeptide sequences unique to the virus. The immunological relevance of the viral unique oligopeptides was assessed by searching them in the immune epitope database (IEDB). We found that HTLV-1 has 15 peptide stretches each consisting of uniquely viral non-human pentapeptides which are ideal candidate for a safe and effective anti-HTLV-1 vaccine. Indeed, experimentally validated HTLV-1 epitopes, as retrieved from the IEDB, contain peptide sequences also present in a vast number of human proteins, thus potentially instituting the basis for cross-reactions. We found a potential for cross-reactivity between the virus and the human proteome and described an epitope platform to be used in order to avoid it, thus obtaining effective, specific, and safe immunization. Potential advantages for mRNA and peptide-based vaccine formulations are discussed.


Subject(s)
Epitopes/chemistry , HTLV-I Infections/prevention & control , Human T-lymphotropic virus 1/immunology , RNA, Messenger/chemistry , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , mRNA Vaccines/immunology , Amino Acid Sequence , Databases, Genetic , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , HTLV-I Infections/immunology , HTLV-I Infections/virology , Human T-lymphotropic virus 1/chemistry , Human T-lymphotropic virus 1/genetics , Humans , Immunization , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics , Viral Vaccines/chemistry , Viral Vaccines/genetics , mRNA Vaccines/chemistry , mRNA Vaccines/genetics
14.
Mol Ther ; 29(9): 2782-2793, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34058388

ABSTRACT

We present a live-attenuated RNA hybrid vaccine technology that uses an RNA vaccine delivery vehicle to deliver in vitro-transcribed, full-length, live-attenuated viral genomes to the site of vaccination. This technology allows ready manufacturing in a cell-free environment, regardless of viral attenuation level, and it promises to avoid many safety and manufacturing challenges of traditional live-attenuated vaccines. We demonstrate this technology through development and testing of a live-attenuated RNA hybrid vaccine against Chikungunya virus (CHIKV), comprised of an in vitro-transcribed, highly attenuated CHIKV genome delivered by a highly stable nanostructured lipid carrier (NLC) formulation as an intramuscular injection. We demonstrate that single-dose immunization of immunocompetent C57BL/6 mice results in induction of high CHIKV-neutralizing antibody titers and protection against mortality and footpad swelling after lethal CHIKV challenge.


Subject(s)
Antibodies, Neutralizing/blood , Chikungunya Fever/prevention & control , Chikungunya virus/genetics , Lipids/chemistry , mRNA Vaccines/administration & dosage , Animals , Antibodies, Viral/blood , Chikungunya Fever/immunology , Chikungunya virus/immunology , Chlorocebus aethiops , Disease Models, Animal , Drug Compounding , Female , Genome, Viral , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nanostructures , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/immunology , mRNA Vaccines/chemistry , mRNA Vaccines/immunology
15.
Mol Ther ; 29(9): 2769-2781, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33992803

ABSTRACT

It is well established that memory CD8 T cells protect susceptible strains of mice from mousepox, a lethal viral disease caused by ectromelia virus (ECTV), the murine counterpart to human variola virus. While mRNA vaccines induce protective antibody (Ab) responses, it is unknown whether they also induce protective memory CD8 T cells. We now show that immunization with different doses of unmodified or N(1)-methylpseudouridine-modified mRNA (modified mRNA) in lipid nanoparticles (LNP) encoding the ECTV gene EVM158 induced similarly strong CD8 T cell responses to the epitope TSYKFESV, albeit unmodified mRNA-LNP had adverse effects at the inoculation site. A single immunization with 10 µg modified mRNA-LNP protected most susceptible mice from mousepox, and booster vaccination increased the memory CD8 T cell pool, providing full protection. Moreover, modified mRNA-LNP encoding TSYKFESV appended to green fluorescent protein (GFP) protected against wild-type ECTV infection while lymphocytic choriomeningitis virus glycoprotein (GP) modified mRNA-LNP protected against ECTV expressing GP epitopes. Thus, modified mRNA-LNP can be used to create protective CD8 T cell-based vaccines against viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Viral Proteins/genetics , mRNA Vaccines/administration & dosage , Animals , Drug Compounding , Ectromelia, Infectious/immunology , Immunization, Secondary , Immunologic Memory , Liposomes , Male , Mice , Nanoparticles , Peptides/chemistry , Peptides/genetics , Peptides/immunology , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/pharmacology , mRNA Vaccines/chemistry , mRNA Vaccines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...