Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
2.
J Med Virol ; 96(6): e29739, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38899449

ABSTRACT

This longitudinal prospective controlled multicenter study aimed to monitor immunity generated by three exposures caused by breakthrough infections (BTI) after COVID-19-vaccination considering pre-existing cell-mediated immunity to common-corona-viruses (CoV) which may impact cellular reactivity against SARS-CoV-2. Anti-SARS-CoV-2-spike-IgG antibodies (anti-S-IgG) and cellular reactivity against Spike-(S)- and nucleocapsid-(N)-proteins were determined in fully-vaccinated (F) individuals who either experienced BTI (F+BTI) or had booster vaccination (F+Booster) compared to partially vaccinated (P+BTI) and unvaccinated (U) from 1 to 24 weeks post PCR-confirmed infection. High avidity anti-S-IgG were found in F+BTI compared to U, the latter exhibiting increased long-lasting pro-inflammatory cytokines to S-stimulation. CoV was associated with higher cellular reactivity in U, whereas no association was seen in F. The study illustrates the induction of significant S-specific cellular responses in F+BTI building-up basic immunity by three exposures. Only U seem to benefit from pre-existing CoV immunity but demonstrated inflammatory immune responses compared to F+BTI who immunologically benefit from enhanced humoral and cellular immunity after BTI. This study demonstrates that individuals with hybrid immunity from COVID-19-vaccination and BTI acquire a stable humoral and cellular immune response that is maintained for at least 6 months. Our findings corroborate recommendations by health authorities to build on basic immunity by three S-protein exposures.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunity, Cellular , Immunoglobulin G , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19/immunology , COVID-19/prevention & control , Antibodies, Viral/blood , Antibodies, Viral/immunology , Male , Female , SARS-CoV-2/immunology , Middle Aged , Adult , Prospective Studies , Spike Glycoprotein, Coronavirus/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunoglobulin G/blood , Longitudinal Studies , Vaccination , Phosphoproteins/immunology , Coronavirus Nucleocapsid Proteins/immunology , Aged , Immunization, Secondary , Cytokines/immunology , BNT162 Vaccine/immunology , BNT162 Vaccine/administration & dosage , 2019-nCoV Vaccine mRNA-1273/immunology , mRNA Vaccines/immunology , Breakthrough Infections
3.
Pharmacol Res Perspect ; 12(3): e1218, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867495

ABSTRACT

According to the CDC, both Pfizer and Moderna COVID-19 vaccines contain nucleoside-modified messenger RNA (mRNA) encoding the viral spike glycoprotein of severe acute respiratory syndrome caused by corona virus (SARS-CoV-2), administered via intramuscular injections. Despite their worldwide use, very little is known about how nucleoside modifications in mRNA sequences affect their breakdown, transcription and protein synthesis. It was hoped that resident and circulating immune cells attracted to the injection site make copies of the spike protein while the injected mRNA degrades within a few days. It was also originally estimated that recombinant spike proteins generated by mRNA vaccines would persist in the body for a few weeks. In reality, clinical studies now report that modified SARS-CoV-2 mRNA routinely persist up to a month from injection and can be detected in cardiac and skeletal muscle at sites of inflammation and fibrosis, while the recombinant spike protein may persist a little over half a year in blood. Vaccination with 1-methylΨ (pseudouridine enriched) mRNA can elicit cellular immunity to peptide antigens produced by +1 ribosomal frameshifting in major histocompatibility complex-diverse people. The translation of 1-methylΨ mRNA using liquid chromatography tandem mass spectrometry identified nine peptides derived from the mRNA +1 frame. These products impact on off-target host T cell immunity that include increased production of new B cell antigens with far reaching clinical consequences. As an example, a highly significant increase in heart muscle 18-flourodeoxyglucose uptake was detected in vaccinated patients up to half a year (180 days). This review article focuses on medical biochemistry, proteomics and deutenomics principles that explain the persisting spike phenomenon in circulation with organ-related functional damage even in asymptomatic individuals. Proline and hydroxyproline residues emerge as prominent deuterium (heavy hydrogen) binding sites in structural proteins with robust isotopic stability that resists not only enzymatic breakdown, but virtually all (non)-enzymatic cleavage mechanisms known in chemistry.


Subject(s)
COVID-19 Vaccines , COVID-19 , RNA, Messenger , Spike Glycoprotein, Coronavirus , Humans , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/immunology , mRNA Vaccines/immunology , Pseudouridine , Recombinant Proteins/administration & dosage , RNA, Viral , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Vaccination , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage
4.
Nature ; 630(8018): 950-960, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749479

ABSTRACT

Immune imprinting is a phenomenon in which prior antigenic experiences influence responses to subsequent infection or vaccination1,2. The effects of immune imprinting on serum antibody responses after boosting with variant-matched SARS-CoV-2 vaccines remain uncertain. Here we characterized the serum antibody responses after mRNA vaccine boosting of mice and human clinical trial participants. In mice, a single dose of a preclinical version of mRNA-1273 vaccine encoding Wuhan-1 spike protein minimally imprinted serum responses elicited by Omicron boosters, enabling generation of type-specific antibodies. However, imprinting was observed in mice receiving an Omicron booster after two priming doses of mRNA-1273, an effect that was mitigated by a second booster dose of Omicron vaccine. In both SARS-CoV-2-infected and uninfected humans who received two Omicron-matched boosters after two or more doses of the prototype mRNA-1273 vaccine, spike-binding and neutralizing serum antibodies cross-reacted with Omicron variants as well as more distantly related sarbecoviruses. Because serum neutralizing responses against Omicron strains and other sarbecoviruses were abrogated after pre-clearing with Wuhan-1 spike protein, antibodies induced by XBB.1.5 boosting in humans focus on conserved epitopes targeted by the antecedent mRNA-1273 primary series. Thus, the antibody response to Omicron-based boosters in humans is imprinted by immunizations with historical mRNA-1273 vaccines, but this outcome may be beneficial as it drives expansion of cross-neutralizing antibodies that inhibit infection of emerging SARS-CoV-2 variants and distantly related sarbecoviruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , mRNA Vaccines , Adult , Animals , Female , Humans , Male , Mice , 2019-nCoV Vaccine mRNA-1273/administration & dosage , 2019-nCoV Vaccine mRNA-1273/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Antibodies, Viral/immunology , Antibodies, Viral/blood , China , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Cross Reactions/immunology , Epitopes, B-Lymphocyte/immunology , mRNA Vaccines/administration & dosage , mRNA Vaccines/genetics , mRNA Vaccines/immunology , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Vaccination
5.
STAR Protoc ; 5(2): 103087, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38795353

ABSTRACT

Here, we present a protocol for the development of mRNA-loaded lipid nanoparticle (LNP) vaccines for target antigen sequences of interest. We describe key steps required to design and synthesize mRNA constructs, their LNP encapsulation, and mouse immunization. We then detail quality control assays to determine RNA purity, guidelines to measure RNA immunogenicity using in vitro reporter systems, and a technique to evaluate antigen-specific T cell responses following immunization.


Subject(s)
Immunization , Lipids , Nanoparticles , RNA, Messenger , Animals , Mice , Nanoparticles/chemistry , RNA, Messenger/genetics , Lipids/chemistry , Immunization/methods , Vaccines/immunology , Vaccines/chemistry , Vaccines/administration & dosage , mRNA Vaccines/immunology , Nanovaccines , Liposomes
6.
Virology ; 596: 110125, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38805804

ABSTRACT

Influenza viruses present a significant threat to global health. The production of a universal vaccine is considered essential due to the ineffectiveness of current seasonal influenza vaccines against mutant strains. mRNA technology offers new prospects in vaccinology, with various candidates for different infectious diseases currently in development and testing phases. In this study, we encapsulated a universal influenza mRNA vaccine. The vaccine encoded influenza hemagglutinin (HA), nucleoprotein (NP), and three tandem repeats of matrix protein 2 (3M2e). Twice-vaccinated mice exhibited strong humoral and cell-mediated immune responses in vivo. Notably, these immune responses led to a significant reduction in viral load of the lungs in challenged mice, and also conferred protection against future wild-type H1N1, H3N2, or H5N1 influenza virus challenges. Our findings suggest that this mRNA-universal vaccine strategy for influenza virus may be instrumental in mitigating the impact of future influenza pandemics.


Subject(s)
Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Viral Matrix Proteins , mRNA Vaccines , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Antibodies, Viral/immunology , mRNA Vaccines/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Female , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Cross Protection/immunology , Viral Load , Lung/virology , Lung/immunology , Humans , Viroporin Proteins
7.
Cell Rep ; 43(6): 114269, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38787725

ABSTRACT

The 2022 mpox outbreak led the World Health Organization (WHO) to declare it a public health emergency of international concern (PHEIC). There is a need to develop more effective and safer mpox virus (MPXV)-specific vaccines in response to the mpox epidemic. The mRNA vaccine is a promising platform to protect against MPXV infection. In this study, we construct two bivalent MPXV mRNA vaccines, designated LBA (B6R-A29L) and LAM (A35R-M1R), and a quadrivalent mRNA vaccine, LBAAM (B6R-A35R-A29L-M1R). The immunogenicity and protective efficacy of these vaccines alone or in combination were evaluated in a lethal mouse model. All mRNA vaccine candidates could elicit potential antigen-specific humoral and cellular immune responses and provide protection against vaccinia virus (VACV) infection. The protective effect of the combination of two bivalent mRNA vaccines and the quadrivalent vaccine was superior to that of the individual bivalent mRNA vaccine. Our study provides valuable insights for the development of more efficient and safer mRNA vaccines against mpox.


Subject(s)
Vaccinia virus , mRNA Vaccines , Animals , Vaccinia virus/immunology , Vaccinia virus/genetics , Mice , Female , mRNA Vaccines/immunology , Humans , Mice, Inbred BALB C , Mpox (monkeypox)/prevention & control , Mpox (monkeypox)/immunology , Vaccinia/immunology , Vaccinia/prevention & control , Antibodies, Viral/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Immunity, Humoral
8.
JCI Insight ; 9(9)2024 May 08.
Article in English | MEDLINE | ID: mdl-38716734

ABSTRACT

mRNA vaccines are likely to become widely used for the prevention of infectious diseases in the future. Nevertheless, a notable gap exists in mechanistic data, particularly concerning the potential effects of sequential mRNA immunization or preexisting immunity on the early innate immune response triggered by vaccination. In this study, healthy adults, with or without documented prior SARS-CoV-2 infection, were vaccinated with the BNT162b2/Comirnaty mRNA vaccine. Prior infection conferred significantly stronger induction of proinflammatory and type I IFN-related gene signatures, serum cytokines, and monocyte expansion after the prime vaccination. The response to the second vaccination further increased the magnitude of the early innate response in both study groups. The third vaccination did not further increase vaccine-induced inflammation. In vitro stimulation of PBMCs with TLR ligands showed no difference in cytokine responses between groups, or before or after prime vaccination, indicating absence of a trained immunity effect. We observed that levels of preexisting antigen-specific CD4 T cells, antibody, and memory B cells correlated with elements of the early innate response to the first vaccination. Our data thereby indicate that preexisting memory formed by infection may augment the innate immune activation induced by mRNA vaccines.


Subject(s)
BNT162 Vaccine , COVID-19 Vaccines , COVID-19 , Cytokines , Immunity, Innate , SARS-CoV-2 , Vaccination , Humans , Immunity, Innate/immunology , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Adult , Male , BNT162 Vaccine/immunology , BNT162 Vaccine/administration & dosage , Female , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Vaccination/methods , Cytokines/immunology , mRNA Vaccines/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Middle Aged , CD4-Positive T-Lymphocytes/immunology , Young Adult , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage
9.
Signal Transduct Target Ther ; 9(1): 118, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702343

ABSTRACT

Antitumor therapies based on adoptively transferred T cells or oncolytic viruses have made significant progress in recent years, but the limited efficiency of their infiltration into solid tumors makes it difficult to achieve desired antitumor effects when used alone. In this study, an oncolytic virus (rVSV-LCMVG) that is not prone to induce virus-neutralizing antibodies was designed and combined with adoptively transferred T cells. By transforming the immunosuppressive tumor microenvironment into an immunosensitive one, in B16 tumor-bearing mice, combination therapy showed superior antitumor effects than monotherapy. This occurred whether the OV was administered intratumorally or intravenously. Combination therapy significantly increased cytokine and chemokine levels within tumors and recruited CD8+ T cells to the TME to trigger antitumor immune responses. Pretreatment with adoptively transferred T cells and subsequent oncolytic virotherapy sensitizes refractory tumors by boosting T-cell recruitment, down-regulating the expression of PD-1, and restoring effector T-cell function. To offer a combination therapy with greater translational value, mRNA vaccines were introduced to induce tumor-specific T cells instead of adoptively transferred T cells. The combination of OVs and mRNA vaccine also displays a significant reduction in tumor burden and prolonged survival. This study proposed a rational combination therapy of OVs with adoptive T-cell transfer or mRNA vaccines encoding tumor-associated antigens, in terms of synergistic efficacy and mechanism.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Animals , Mice , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Oncolytic Virotherapy/methods , Combined Modality Therapy , mRNA Vaccines/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Tumor Microenvironment/immunology , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , Humans , Cell Line, Tumor , Cancer Vaccines/immunology , Cancer Vaccines/genetics , Cancer Vaccines/administration & dosage
10.
Cell Chem Biol ; 31(5): 830-832, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759615

ABSTRACT

The rise of immunotherapy and mRNA vaccines has underscored the power of modulating the immune system for a desired response. In this Voices piece, the Cell Chemical Biology editors ask researchers from a range of backgrounds: what are some major challenges and opportunities facing the field in coming years?


Subject(s)
Immune System , Immunotherapy , Humans , Immune System/immunology , Immune System/metabolism , mRNA Vaccines/immunology
11.
Methods Mol Biol ; 2786: 51-87, 2024.
Article in English | MEDLINE | ID: mdl-38814390

ABSTRACT

Vectored RNA vaccines offer a variety of possibilities to engineer targeted vaccines. They are cost-effective and safe, but replication competent, activating the humoral as well as the cellular immune system.This chapter focuses on RNA vaccines derived from negative-strand RNA viruses from the order Mononegavirales with special attention to Newcastle disease virus-based vaccines and their generation. It shall provide an overview on the advantages and disadvantages of certain vector platforms as well as their scopes of application, including an additional section on experimental COVID-19 vaccines.


Subject(s)
Genetic Vectors , Newcastle disease virus , mRNA Vaccines , Animals , Humans , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Genetic Vectors/genetics , Newcastle disease virus/genetics , Newcastle disease virus/immunology , RNA Viruses/genetics , RNA Viruses/immunology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , mRNA Vaccines/genetics , mRNA Vaccines/immunology
12.
Nat Commun ; 15(1): 4350, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782954

ABSTRACT

mRNA lipid nanoparticle (LNP) vaccines would be useful during an influenza virus pandemic since they can be produced rapidly and do not require the generation of egg-adapted vaccine seed stocks. Highly pathogenic avian influenza viruses from H5 clade 2.3.4.4b are circulating at unprecedently high levels in wild and domestic birds and have the potential to adapt to humans. Here, we generate an mRNA lipid nanoparticle (LNP) vaccine encoding the hemagglutinin (HA) glycoprotein from a clade 2.3.4.4b H5 isolate. The H5 mRNA-LNP vaccine elicits strong T cell and antibody responses in female mice, including neutralizing antibodies and broadly-reactive anti-HA stalk antibodies. The H5 mRNA-LNP vaccine elicits antibodies at similar levels compared to whole inactivated vaccines in female mice with and without prior H1N1 exposures. Finally, we find that the H5 mRNA-LNP vaccine is immunogenic in male ferrets and prevents morbidity and mortality of animals following 2.3.4.4b H5N1 challenge. Together, our data demonstrate that a monovalent mRNA-LNP vaccine expressing 2.3.4.4b H5 is immunogenic and protective in pre-clinical animal models.


Subject(s)
Antibodies, Viral , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Nanoparticles , Orthomyxoviridae Infections , mRNA Vaccines , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Female , Mice , Nanoparticles/chemistry , Male , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , mRNA Vaccines/immunology , Antibodies, Neutralizing/immunology , Mice, Inbred BALB C , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Influenza in Birds/virology , Humans , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Birds/virology , Lipids/chemistry , Liposomes
13.
Malar J ; 23(1): 136, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711053

ABSTRACT

Malaria vaccine introduction in endemic countries is a game-changing milestone in the fight against the disease. This article examines the inequity in the global pharmaceutical research, development, manufacturing, and trade landscape. The role of inequity in hindering progress towards malaria elimination is explored. The analysis finds that transformational changes are required to create an equity-enabling environment. Addressing the inequity is critical to maximizing the public health impact of vaccines and attaining sustainability. Avenues to catalyze progress by leveraging malaria vaccines and messenger ribonucleic acid (mRNA) technology are discussed.


Subject(s)
Malaria Vaccines , Malaria , mRNA Vaccines , Humans , Disease Eradication/methods , Global Health , Malaria/immunology , Malaria/prevention & control , Malaria Vaccines/immunology , Malaria Vaccines/genetics , Pharmaceutical Research , mRNA Vaccines/immunology , Africa
14.
Science ; 384(6697): eadk0582, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38753770

ABSTRACT

Germline-targeting (GT) HIV vaccine strategies are predicated on deriving broadly neutralizing antibodies (bnAbs) through multiple boost immunogens. However, as the recruitment of memory B cells (MBCs) to germinal centers (GCs) is inefficient and may be derailed by serum antibody-induced epitope masking, driving further B cell receptor (BCR) modification in GC-experienced B cells after boosting poses a challenge. Using humanized immunoglobulin knockin mice, we found that GT protein trimer immunogen N332-GT5 could prime inferred-germline precursors to the V3-glycan-targeted bnAb BG18 and that B cells primed by N332-GT5 were effectively boosted by either of two novel protein immunogens designed to have minimum cross-reactivity with the off-target V1-binding responses. The delivery of the prime and boost immunogens as messenger RNA lipid nanoparticles (mRNA-LNPs) generated long-lasting GCs, somatic hypermutation, and affinity maturation and may be an effective tool in HIV vaccine development.


Subject(s)
AIDS Vaccines , Broadly Neutralizing Antibodies , Germinal Center , HIV Antibodies , HIV-1 , Immunization, Secondary , Nanoparticles , mRNA Vaccines , Animals , Humans , Mice , AIDS Vaccines/immunology , B-Lymphocytes/immunology , Broadly Neutralizing Antibodies/immunology , Cross Reactions , Gene Knock-In Techniques , Germinal Center/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , HIV-1/genetics , Liposomes , Memory B Cells/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/genetics , Somatic Hypermutation, Immunoglobulin , mRNA Vaccines/immunology , Female , Mice, Inbred C57BL
15.
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
16.
Influenza Other Respir Viruses ; 18(5): e13309, 2024 May.
Article in English | MEDLINE | ID: mdl-38725111

ABSTRACT

BACKGROUND: The newly emerged SARS-CoV-2 possesses shared antigenic epitopes with other human coronaviruses. We investigated if COVID-19 vaccination or SARS-CoV-2 infection may boost cross-reactive antibodies to other human coronaviruses. METHODS: Prevaccination and postvaccination sera from SARS-CoV-2 naïve healthy subjects who received three doses of the mRNA vaccine (BioNTech, BNT) or the inactivated vaccine (CoronaVac, CV) were used to monitor the level of cross-reactive antibodies raised against other human coronaviruses by enzyme-linked immunosorbent assay. In comparison, convalescent sera from COVID-19 patients with or without prior vaccination history were also tested. Pseudoparticle neutralization assay was performed to detect neutralization antibody against MERS-CoV. RESULTS: Among SARS-CoV-2 infection-naïve subjects, BNT or CV significantly increased the anti-S2 antibodies against Betacoronaviruses (OC43 and MERS-CoV) but not Alphacoronaviruses (229E). The prevaccination antibody response to the common cold human coronaviruses did not negatively impact the postvaccination antibody response to SARS-CoV-2. Cross-reactive antibodies that binds to the S2 protein of MERS-CoV were similarly detected from the convalescent sera of COVID-19 patients with or without vaccination history. However, these anti-S2 antibodies do not possess neutralizing activity in MERS-CoV pseudoparticle neutralization tests. CONCLUSIONS: Our results suggest that SARS-CoV-2 infection or vaccination may potentially modulate population immune landscape against previously exposed or novel human coronaviruses. The findings have implications for future sero-epidemiological studies on MERS-CoV.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Cross Reactions , SARS-CoV-2 , Humans , Cross Reactions/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Adult , Male , Female , Vaccination , Middle Aged , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Neutralization Tests , Middle East Respiratory Syndrome Coronavirus/immunology , Young Adult , mRNA Vaccines/immunology
17.
Nat Med ; 30(5): 1363-1372, 2024 May.
Article in English | MEDLINE | ID: mdl-38637636

ABSTRACT

Here we conducted a multicenter open-label, randomized phase 2 and 3 study to assess the safety and immunogenicity of a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron-specific (BA.1/B.1.1.529), monovalent, thermostable, self-amplifying mRNA vaccine, GEMCOVAC-OM, when administered intradermally as a booster in healthy adults who had received two doses of BBV152 or ChAdOx1 nCoV-19. GEMCOVAC-OM was well tolerated with no related serious adverse events in both phase 2 and phase 3. In phase 2, the safety and immunogenicity of GEMCOVAC-OM was compared with our prototype mRNA vaccine GEMCOVAC-19 (D614G variant-specific) in 140 participants. At day 29 after vaccination, there was a significant rise in anti-spike (BA.1) IgG antibodies with GEMCOVAC-OM (P < 0.0001) and GEMCOVAC-19 (P < 0.0001). However, the IgG titers (primary endpoint) and seroconversion were higher with GEMCOVAC-OM (P < 0.0001). In phase 3, GEMCOVAC-OM was compared with ChAdOx1 nCoV-19 in 3,140 participants (safety cohort), which included an immunogenicity cohort of 420 participants. At day 29, neutralizing antibody titers against the BA.1 variant of SARS-CoV-2 were significantly higher than baseline in the GEMCOVAC-OM arm (P < 0.0001), but not in the ChAdOx1 nCoV-19 arm (P = 0.1490). GEMCOVAC-OM was noninferior (primary endpoint) and superior to ChAdOx1 nCoV-19 in terms of neutralizing antibody titers and seroconversion rate (lower bound 95% confidence interval of least square geometric mean ratio >1 and difference in seroconversion >0% for superiority). At day 29, anti-spike IgG antibodies and seroconversion (secondary endpoints) were significantly higher with GEMCOVAC-OM (P < 0.0001). These results demonstrate that GEMCOVAC-OM is safe and boosts immune responses against the B.1.1.529 variant. Clinical Trial Registry India identifier: CTRI/2022/10/046475 .


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/adverse effects , SARS-CoV-2/immunology , Male , Female , Adult , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Middle Aged , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Immunoglobulin G/immunology , Immunoglobulin G/blood , Young Adult , mRNA Vaccines/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Immunogenicity, Vaccine/immunology , ChAdOx1 nCoV-19/immunology
18.
Elife ; 132024 Apr 24.
Article in English | MEDLINE | ID: mdl-38656290

ABSTRACT

Background: End-stage renal disease (ESRD) patients experience immune compromise characterized by complex alterations of both innate and adaptive immunity, and results in higher susceptibility to infection and lower response to vaccination. This immune compromise, coupled with greater risk of exposure to infectious disease at hemodialysis (HD) centers, underscores the need for examination of the immune response to the COVID-19 mRNA-based vaccines. Methods: The immune response to the COVID-19 BNT162b2 mRNA vaccine was assessed in 20 HD patients and cohort-matched controls. RNA sequencing of peripheral blood mononuclear cells was performed longitudinally before and after each vaccination dose for a total of six time points per subject. Anti-spike antibody levels were quantified prior to the first vaccination dose (V1D0) and 7 d after the second dose (V2D7) using anti-spike IgG titers and antibody neutralization assays. Anti-spike IgG titers were additionally quantified 6 mo after initial vaccination. Clinical history and lab values in HD patients were obtained to identify predictors of vaccination response. Results: Transcriptomic analyses demonstrated differing time courses of immune responses, with prolonged myeloid cell activity in HD at 1 wk after the first vaccination dose. HD also demonstrated decreased metabolic activity and decreased antigen presentation compared to controls after the second vaccination dose. Anti-spike IgG titers and neutralizing function were substantially elevated in both controls and HD at V2D7, with a small but significant reduction in titers in HD groups (p<0.05). Anti-spike IgG remained elevated above baseline at 6 mo in both subject groups. Anti-spike IgG titers at V2D7 were highly predictive of 6-month titer levels. Transcriptomic biomarkers after the second vaccination dose and clinical biomarkers including ferritin levels were found to be predictive of antibody development. Conclusions: Overall, we demonstrate differing time courses of immune responses to the BTN162b2 mRNA COVID-19 vaccination in maintenance HD subjects comparable to healthy controls and identify transcriptomic and clinical predictors of anti-spike IgG titers in HD. Analyzing vaccination as an in vivo perturbation, our results warrant further characterization of the immune dysregulation of ESRD. Funding: F30HD102093, F30HL151182, T32HL144909, R01HL138628. This research has been funded by the University of Illinois at Chicago Center for Clinical and Translational Science (CCTS) award UL1TR002003.


Subject(s)
Antibodies, Viral , BNT162 Vaccine , COVID-19 Vaccines , COVID-19 , Kidney Failure, Chronic , Renal Dialysis , SARS-CoV-2 , Humans , Male , Female , Middle Aged , COVID-19/immunology , COVID-19/prevention & control , BNT162 Vaccine/immunology , BNT162 Vaccine/administration & dosage , Aged , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Viral/blood , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Kidney Failure, Chronic/immunology , Transcriptome , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Immunoglobulin G/blood , mRNA Vaccines/immunology , Vaccination
19.
Int Immunopharmacol ; 132: 112037, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38599100

ABSTRACT

Colorectal cancer (CRC) is a typical cancer that accounts for 10% of all new cancer cases annually and nearly 10% of all cancer deaths. Despite significant progress in current classical interventions for CRC, these traditional strategies could be invasive and with numerous adverse effects. The poor prognosis of CRC patients highlights the evident and pressing need for more efficient and targeted treatment. Novel strategies regarding mRNA vaccines for anti-tumor therapy have also been well-developed since the successful application for the prevention of COVID-19. mRNA vaccine technology won the 2023 Nobel Prize in Physiology or Medicine, signaling a new direction in human anti-cancer treatment: mRNA medicine. As a promising new immunotherapy in CRC and other multiple cancer treatments, the mRNA vaccine has higher specificity, better efficacy, and fewer side effects than traditional strategies. The present review outlines the basics of mRNA vaccines and their advantages over other vaccines and informs an available strategy for developing efficient mRNA vaccines for CRC precise treatment. In the future, more exploration of mRNA vaccines for CRC shall be attached, fostering innovation to address existing limitations.


Subject(s)
Cancer Vaccines , Colorectal Neoplasms , Immunotherapy , mRNA Vaccines , Animals , Humans , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Immunotherapy/methods , mRNA Vaccines/immunology , mRNA Vaccines/therapeutic use
20.
Mol Ther ; 32(5): 1266-1283, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38569556

ABSTRACT

Carrier-free naked mRNA vaccines may reduce the reactogenicity associated with delivery carriers; however, their effectiveness against infectious diseases has been suboptimal. To boost efficacy, we targeted the skin layer rich in antigen-presenting cells (APCs) and utilized a jet injector. The jet injection efficiently introduced naked mRNA into skin cells, including APCs in mice. Further analyses indicated that APCs, after taking up antigen mRNA in the skin, migrated to the lymph nodes (LNs) for antigen presentation. Additionally, the jet injection provoked localized lymphocyte infiltration in the skin, serving as a physical adjuvant for vaccination. Without a delivery carrier, our approach confined mRNA distribution to the injection site, preventing systemic mRNA leakage and associated systemic proinflammatory reactions. In mouse vaccination, the naked mRNA jet injection elicited robust antigen-specific antibody production over 6 months, along with germinal center formation in LNs and the induction of both CD4- and CD8-positive T cells. By targeting the SARS-CoV-2 spike protein, this approach provided protection against viral challenge. Furthermore, our approach generated neutralizing antibodies against SARS-CoV-2 in non-human primates at levels comparable to those observed in mice. In conclusion, our approach offers a safe and effective option for mRNA vaccines targeting infectious diseases.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , mRNA Vaccines , Animals , Mice , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , mRNA Vaccines/immunology , COVID-19/prevention & control , COVID-19/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral/immunology , Female , Antigen-Presenting Cells/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , CD8-Positive T-Lymphocytes/immunology , Antibodies, Neutralizing/immunology , Humans , Vaccination/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...