Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.150
Filter
1.
PLoS Pathog ; 20(9): e1012508, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39303003

ABSTRACT

Influenza and coronavirus disease 2019 (COVID-19) represent two respiratory diseases that have significantly impacted global health, resulting in substantial disease burden and mortality. An optimal solution would be a combined vaccine capable of addressing both diseases, thereby obviating the need for multiple vaccinations. Previously, we conceived a chimeric protein subunit vaccine targeting both influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), utilizing the receptor binding domain of spike protein (S-RBD) and the stalk region of hemagglutinin protein (HA-stalk) components. By integrating the S-RBD from the SARS-CoV-2 Delta variant with the headless hemagglutinin (HA) from H1N1 influenza virus, we constructed stable trimeric structures that remain accessible to neutralizing antibodies. This vaccine has demonstrated its potential by conferring protection against a spectrum of strains in mouse models. In this study, we designed an mRNA vaccine candidate encoding the chimeric antigen. The resultant humoral and cellular immune responses were meticulously evaluated in mouse models. Furthermore, the protective efficacy of the vaccine was rigorously examined through challenges with either homologous or heterologous influenza viruses or SARS-CoV-2 strains. Our findings reveal that the mRNA vaccine exhibited robust immunogenicity, engendering high and sustained levels of neutralizing antibodies accompanied by robust and persistent cellular immunity. Notably, this vaccine effectively afforded complete protection to mice against H1N1 or heterosubtypic H5N8 subtypes, as well as the SARS-CoV-2 Delta and Omicron BA.2 variants. Additionally, our mRNA vaccine design can be easily adapted from Delta RBD to Omicron RBD antigens, providing protection against emerging variants. The development of two-in-one vaccine targeting both influenza and COVID-19, incorporating the mRNA platform, may provide a versatile approach to combating future pandemics.


Subject(s)
COVID-19 Vaccines , COVID-19 , Hemagglutinin Glycoproteins, Influenza Virus , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , mRNA Vaccines , Animals , Mice , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19/immunology , mRNA Vaccines/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Humans , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , COVID-19 Vaccines/immunology , Influenza Vaccines/immunology , Antibodies, Viral/immunology , Mice, Inbred BALB C , Female , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Vaccines, Synthetic/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Antibodies, Neutralizing/immunology
2.
Front Immunol ; 15: 1433442, 2024.
Article in English | MEDLINE | ID: mdl-39295862

ABSTRACT

The monoclonal antibody rituximab improves clinical outcome in the treatment of CD20-positive lymphomatous neoplasms, and it is an established drug for treatment of these cancers. Successful mRNA COVID-19 (SARS-CoV-2) vaccination is extremely important for lymphoma patients because they tend to be elderly with comorbidities which leaves them at increased risk of poor outcomes once infected by Coronavirus. Anti-CD20 therapies such as rituximab, deplete B-cell populations and can affect vaccine efficacy. Therefore, a knowledge of the effect of COVID-19 vaccination in this group is critical. We followed a cohort of 28 patients with CD20-positive lymphomatous malignancies treated with rituximab that started prior to their course of COVID-19 vaccination, including boosters. We assayed for vaccine "take" in the humoral (IgG and IgA) and cellular compartment. Here, we show that short-term and long-term development of IgG and IgA antibodies directed toward COVID-19 spike protein are reduced in these patients compared to healthy controls. Conversely, the robustness and breath of underlying T-cell response is equal to healthy controls. This response is not limited to specific parts of the spike protein but spans the spike region, including response to the conserved Receptor Binding Domain (RBD). Our data informs on rational vaccine design and bodes well for future vaccination strategies that require strong induction of T-cell responses in these patients.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Lymphoma , Rituximab , SARS-CoV-2 , Humans , Rituximab/therapeutic use , COVID-19/immunology , COVID-19/prevention & control , Female , Male , Aged , SARS-CoV-2/immunology , Middle Aged , COVID-19 Vaccines/immunology , Lymphoma/immunology , Lymphoma/drug therapy , Lymphoma/therapy , Antibodies, Viral/immunology , Antibodies, Viral/blood , Spike Glycoprotein, Coronavirus/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Immunoglobulin A/immunology , Immunoglobulin A/blood , Antigens, CD20/immunology , Aged, 80 and over , Vaccination , mRNA Vaccines
3.
Oncol Res ; 32(10): 1543-1564, 2024.
Article in English | MEDLINE | ID: mdl-39308511

ABSTRACT

The advent of RNA therapy, particularly through the development of mRNA cancer vaccines, has ushered in a new era in the field of oncology. This article provides a concise overview of the key principles, recent advancements, and potential implications of mRNA cancer vaccines as a groundbreaking modality in cancer treatment. mRNA cancer vaccines represent a revolutionary approach to combatting cancer by leveraging the body's innate immune system. These vaccines are designed to deliver specific mRNA sequences encoding cancer-associated antigens, prompting the immune system to recognize and mount a targeted response against malignant cells. This personalized and adaptive nature of mRNA vaccines holds immense potential for addressing the heterogeneity of cancer and tailoring treatments to individual patients. Recent breakthroughs in the development of mRNA vaccines, exemplified by the success of COVID-19 vaccines, have accelerated their application in oncology. The mRNA platform's versatility allows for the rapid adaptation of vaccine candidates to various cancer types, presenting an agile and promising avenue for therapeutic intervention. Clinical trials of mRNA cancer vaccines have demonstrated encouraging results in terms of safety, immunogenicity, and efficacy. Pioneering candidates, such as BioNTech's BNT111 and Moderna's mRNA-4157, have exhibited promising outcomes in targeting melanoma and solid tumors, respectively. These successes underscore the potential of mRNA vaccines to elicit robust and durable anti-cancer immune responses. While the field holds great promise, challenges such as manufacturing complexities and cost considerations need to be addressed for widespread adoption. The development of scalable and cost-effective manufacturing processes, along with ongoing clinical research, will be pivotal in realizing the full potential of mRNA cancer vaccines. Overall, mRNA cancer vaccines represent a cutting-edge therapeutic approach that holds the promise of transforming cancer treatment. As research progresses, addressing challenges and refining manufacturing processes will be crucial in advancing these vaccines from clinical trials to mainstream oncology practice, offering new hope for patients in the fight against cancer.


Subject(s)
Cancer Vaccines , Neoplasms , Vaccine Development , mRNA Vaccines , Humans , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/prevention & control , COVID-19 Vaccines/immunology , COVID-19/prevention & control , COVID-19/immunology , Vaccines, Synthetic/immunology , RNA, Messenger/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , SARS-CoV-2/immunology , SARS-CoV-2/genetics
4.
Cell ; 187(19): 5171-5194, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39303685

ABSTRACT

The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.


Subject(s)
COVID-19 , Vaccinology , Humans , Vaccinology/methods , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , History, 20th Century , COVID-19 Vaccines/immunology , History, 21st Century , Vaccine Development , SARS-CoV-2/immunology , mRNA Vaccines
5.
Hum Vaccin Immunother ; 20(1): 2399382, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39254005

ABSTRACT

Genetic polymorphisms have been linked to the differential waning of vaccine-induced immunity against COVID-19 following vaccination. Despite this, evidence on the mechanisms behind this waning and its implications for vaccination policy remains limited. We hypothesize that specific gene variants may modulate the development of vaccine-initiated immunity, leading to impaired immune function. This study investigates genetic determinants influencing the sustainability of immunity post-mRNA vaccination through a genome-wide association study (GWAS). Utilizing a hospital-based, test negative case-control design, we enrolled 1,119 participants from the Taiwan Precision Medicine Initiative (TPMI) cohort, all of whom completed a full mRNA COVID-19 vaccination regimen and underwent PCR testing during the Omicron outbreak. Participants were classified into breakthrough and protected groups based on PCR results. Genetic samples were analyzed using SNP arrays with rigorous quality control. Cox regression identified significant single nucleotide polymorphisms (SNPs) associated with breakthrough infections, affecting 743 genes involved in processes such as antigenic protein translation, B cell activation, and T cell function. Key genes identified include CD247, TRPV1, MYH9, CCL16, and RPTOR, which are vital for immune responses. Polygenic risk score (PRS) analysis revealed that individuals with higher PRS are at greater risk of breakthrough infections post-vaccination, demonstrating a high predictability (AUC = 0.787) in validating population. This finding confirms the significant influence of genetic variations on the durability of immune responses and vaccine effectiveness. This study highlights the importance of considering genetic polymorphisms in evaluating vaccine-induced immunity and proposes potential personalized vaccination strategies by tailoring regimens to individual genetic profiles.


Subject(s)
COVID-19 Vaccines , COVID-19 , Genome-Wide Association Study , Polymorphism, Single Nucleotide , SARS-CoV-2 , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , Male , Female , Middle Aged , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Case-Control Studies , Adult , Vaccine Efficacy , Aged , mRNA Vaccines , Taiwan , Vaccination , Precision Medicine , Breakthrough Infections
7.
Sci Rep ; 14(1): 20425, 2024 09 03.
Article in English | MEDLINE | ID: mdl-39227428

ABSTRACT

This study focused on the challenges posed by the fluctuating demand for COVID-19 vaccines, considering factors such as side effects, religious objections, and absenteeism, which result in the accumulation of excess vaccines. Recognizing the resulting social, economic, and environmental issues, this study investigated the application of a lateral transshipment policy for the management of the inventory of short-term vaccines, considering related unpredictabilities. A discrete event simulation built on foundational principles derived from a mixed-integer linear programming model was employed to explore the dynamics of mRNA-based vaccine distribution among two hospitals based on lateral transshipment and reordering policies. Through the simulation of various scenarios over periods of 1-30 days, transshipment based on the availability policy is employed to determine the quantity of vaccines to be transshipped, constrained to vial amounts, and the (s, S) inventory system for reordering. The results of this study underscore the efficacy of lateral transshipment, particularly in situations where demand discrepancies exist between hospitals, thereby revealing its superiority over non-transshipment strategies within 7 days.


Subject(s)
COVID-19 Vaccines , COVID-19 , mRNA Vaccines , Humans , COVID-19 Vaccines/economics , COVID-19/prevention & control , COVID-19/epidemiology , COVID-19/economics , SARS-CoV-2 , Vaccines, Synthetic/economics
8.
Nat Commun ; 15(1): 7745, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39232036

ABSTRACT

Myocarditis is the most salient serious adverse event following messenger RNA-based Covid-19 vaccines. The highest risk is observed after the second dose compared to the first, whereas the level of risk associated with more distant booster doses seems to lie in between. We aimed to assess the relation between dosing interval and the risk of myocarditis, for both the two-dose primary series and the third dose (first booster). This matched case-control study included 7911 cases of myocarditis aged 12 or more in a period where approximately 130 million vaccine doses were administered. Here we show that longer intervals between each consecutive dose, including booster, may decrease the occurrence of vaccine-associated myocarditis by up to a factor of 4, especially under age 50. These results suggest that a minimum 6-month interval might be required when scheduling additional booster vaccination.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunization, Secondary , Myocarditis , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Young Adult , Case-Control Studies , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Immunization Schedule , mRNA Vaccines/adverse effects , Myocarditis/prevention & control , Myocarditis/etiology , Vaccination/adverse effects , Vaccination/methods
9.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39240335

ABSTRACT

Predicting the immunogenicity of candidate vaccines in humans remains a challenge. To address this issue, we developed a lymphoid organ-chip (LO chip) model based on a microfluidic chip seeded with human PBMC at high density within a 3D collagen matrix. Perfusion of the SARS-CoV-2 spike protein mimicked a vaccine boost by inducing a massive amplification of spike-specific memory B cells, plasmablast differentiation, and spike-specific antibody secretion. Features of lymphoid tissue, including the formation of activated CD4+ T cell/B cell clusters and the emigration of matured plasmablasts, were recapitulated in the LO chip. Importantly, myeloid cells were competent at capturing and expressing mRNA vectored by lipid nanoparticles, enabling the assessment of responses to mRNA vaccines. Comparison of on-chip responses to Wuhan monovalent and Wuhan/Omicron bivalent mRNA vaccine boosts showed equivalent induction of Omicron neutralizing antibodies, pointing at immune imprinting as reported in vivo. The LO chip thus represents a versatile platform suited to the preclinical evaluation of vaccine-boosting strategies.


Subject(s)
COVID-19 Vaccines , COVID-19 , Memory B Cells , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , mRNA Vaccines , Humans , COVID-19 Vaccines/immunology , mRNA Vaccines/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Memory B Cells/immunology , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Lymphoid Tissue/immunology , Lab-On-A-Chip Devices , Vaccines, Synthetic/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Liposomes , Nanoparticles
10.
PLoS One ; 19(9): e0307877, 2024.
Article in English | MEDLINE | ID: mdl-39240891

ABSTRACT

Tuberculosis(TB) of the Central nervous system (CNS) is a rare and highly destructive disease. The emergence of drug resistance has increased treatment difficulty, leaving the Bacillus Calmette-Guérin (BCG) vaccine as the only licensed preventative immunization available. This study focused on identifying the epitopes of PknD (Rv0931c) and Rv0986 from Mycobacterium tuberculosis(Mtb) strain H37Rv using an in silico method. The goal was to develop a therapeutic mRNA vaccine for preventing CNS TB. The vaccine was designed to be non-allergenic, non-toxic, and highly antigenic. Codon optimization was performed to ensure effective translation in the human host. Additionally, the secondary and tertiary structures of the vaccine were predicted, and molecular docking with TLR-4 was carried out. A molecular dynamics simulation confirmed the stability of the complex. The results indicate that the vaccine structure shows effectiveness. Overall, the constructed vaccine exhibits ideal physicochemical properties, immune response, and stability, laying a theoretical foundation for future laboratory experiments.


Subject(s)
Computer Simulation , Molecular Docking Simulation , Molecular Dynamics Simulation , Mycobacterium tuberculosis , Tuberculosis, Central Nervous System , Humans , Mycobacterium tuberculosis/immunology , Tuberculosis, Central Nervous System/prevention & control , Tuberculosis, Central Nervous System/immunology , Tuberculosis Vaccines/immunology , Epitopes/immunology , Epitopes/chemistry , mRNA Vaccines , Vaccines, Synthetic/immunology
11.
Vaccine ; 42(25): 126275, 2024 Nov 14.
Article in English | MEDLINE | ID: mdl-39241318

ABSTRACT

BACKGROUND: BBV152 (Covaxin™) is a whole-virion inactivated SARS-CoV-2 vaccine mixed with an immune adjuvant. We aimed to compare immune responses after booster vaccination with heterologous BBV152 versus homologous mRNA vaccine. METHODS: We conducted a randomized, participant-blinded, controlled trial. Fifty mRNA-vaccinated participants were enrolled and randomized to receive an mRNA booster (n = 26) or BBV152 (n = 24). Blood samples were collected pre-vaccination, and at Day 7, 28, 180 and 360 post-booster for analysis of humoral and cellular immune responses. Primary end point was the SARS-CoV-2 anti-spike antibody titer at day 28. RESULTS: Recruitment began in January 2022 and was terminated early due to the BBV152 group meeting pre-specified criteria for futility. At Day 28 post-boost, mean SARS-CoV-2 spike antibody titers were lower with BBV152 (2004 IU/mL; 95 % confidence interval [CI], 1132-3548) vs mRNA (26,669 IU/mL; 95 % CI, 21,330-33,266; p < 0.0001), but comparable levels of spike-specific CD4 and cytotoxic T-cells were observed. Anti-spike antibody titers remained significantly different at Day 180: BBV152 4467 IU/mL (95 % CI, 1959-10,186) vs mRNA 20,749 IU/mL (95 % CI, 12,303-35,075; p = 0.0017). Levels of surrogate virus neutralizing antibodies against ancestral and Omicron subvariants BA.1 and BA.2 were significantly higher among mRNA recipients at Day 180, including after adjusting for intercurrent infection. By Day 360, anti-spike antibody titers and neutralizing antibody levels against Omicron subvariants became similar between vaccine groups. By the end of the study, 16 in each arm (mRNA 64 % and BBV152 69.6 %) had breakthrough infections and time to COVID-19 infection between vaccine groups were similar (p = 0.63). CONCLUSIONS: Wild-type SARS-CoV-2 anti-spike antibody titer and surrogate virus neutralizing test levels against wild-type SARS-CoV-2 and Omicron subvariants BA.1/BA.2/BA.5 were significantly higher at Day 28 and 180 in individuals who received booster vaccination with an mRNA vaccine compared with BBV152. CLINICAL TRIAL REGISTRATION NUMBER: NCT05142319.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , Immunogenicity, Vaccine , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Female , Male , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/prevention & control , COVID-19/immunology , Adult , Immunization, Secondary/methods , Middle Aged , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , mRNA Vaccines/immunology , Young Adult , Immunity, Humoral , Immunity, Cellular , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage
12.
J Biomed Sci ; 31(1): 89, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256822

ABSTRACT

Realizing the immense clinical potential of mRNA-based drugs will require continued development of methods to safely deliver the bioactive agents with high efficiency and without triggering side effects. In this regard, lipid nanoparticles have been successfully utilized to improve mRNA delivery and protect the cargo from extracellular degradation. Encapsulation in lipid nanoparticles was an essential factor in the successful clinical application of mRNA vaccines, which conclusively demonstrated the technology's potential to yield approved medicines. In this review, we begin by describing current advances in mRNA modifications, design of novel lipids and development of lipid nanoparticle components for mRNA-based drugs. Then, we summarize key points pertaining to preclinical and clinical development of mRNA therapeutics. Finally, we cover topics related to targeted delivery systems, including endosomal escape and targeting of immune cells, tumors and organs for use with mRNA vaccines and new treatment modalities for human diseases.


Subject(s)
Drug Delivery Systems , Nanoparticles , RNA, Messenger , Humans , RNA, Messenger/genetics , RNA, Messenger/administration & dosage , Nanoparticles/chemistry , Drug Delivery Systems/methods , mRNA Vaccines , Lipids/chemistry , Liposomes
13.
Adv Exp Med Biol ; 1457: 165-184, 2024.
Article in English | MEDLINE | ID: mdl-39283426

ABSTRACT

In the wake of the novel Coronavirus, it has become imperative to develop vaccines that can alert our immune system to the virus, hence reducing the severity of disease if exposed to it. Different types of vaccines have been studied in this respect, such as nucleic acid vaccines (mRNA and DNA vaccines), vector-based vaccines, whole-virus vaccines (inactivated virus and live-attenuated vaccine), as well as protein subunits vaccines. The results have demonstrated that these vaccines are efficient against both the original strain and emerging variants. Furthermore, they also proved their safety with no grave adverse events. Despite this, hesitancy toward taking these vaccines is still present among certain groups in society due to various factors. Special emphasis has been placed on studies concerning pregnant women, children, elderly people, and immunocompromised individuals where efficacy and safety were proven.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccination , Humans , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Vaccination/methods , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Female , Pregnancy , Immunocompromised Host , mRNA Vaccines/immunology
14.
Front Immunol ; 15: 1442783, 2024.
Article in English | MEDLINE | ID: mdl-39301027

ABSTRACT

Advances in immunotherapy rely on targeting novel cell surface antigens, including therapeutically relevant peptide fragments presented by HLA molecules, collectively known as the actionable immunopeptidome. Although the immunopeptidome of classical HLA molecules is extensively studied, exploration of the peptide repertoire presented by non-classical HLA-E remains limited. Growing evidence suggests that HLA-E molecules present pathogen-derived and tumor-associated peptides to CD8+ T cells, positioning them as promising targets for universal immunotherapies due to their minimal polymorphism. This mini-review highlights recent developments in mass spectrometry (MS) technologies for profiling the HLA-E immunopeptidome in various diseases. We discuss the unique features of HLA-E, its expression patterns, stability, and the potential for identifying new therapeutic targets. Understanding the broad repertoire of actionable peptides presented by HLA-E can lead to innovative treatments for viral and pathogen infections and cancer, leveraging its monomorphic nature for broad therapeutic efficacy.


Subject(s)
HLA-E Antigens , Histocompatibility Antigens Class I , Immunotherapy , Mass Spectrometry , Humans , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Mass Spectrometry/methods , Immunotherapy/methods , mRNA Vaccines , Neoplasms/therapy , Neoplasms/immunology , Peptides/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Antigen Presentation/immunology
15.
Vaccine ; 42(24): 126304, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39236403

ABSTRACT

Repeated exposure of animals to Ixodes scapularis ticks can result in acquired tick resistance (ATR). The first manifestation of ATR is erythema at the tick bite site, however, the specific peptide targets and mechanisms associated with this early aspect of ATR are not understood. In this study, we immunized guinea pigs with a lipid nanoparticle containing the mRNA encoding 25 amino acids in the carboxyl terminus of Salp14 (Salp14-C mRNA-LNP), an I. scapularis salivary protein. The animals produced high titers of IgG directed at the carboxyl terminus of Salp14. Guinea pigs immunized with Salp14-C mRNA-LNP and then exposed to I. scapularis, developed erythema at the tick bite site. Transcriptomics of the skin of guinea pigs at the I. scapularis bite sites elucidated selected pathways, including histamine activation, that are associated with the development of erythema. The study demonstrates that an mRNA vaccine encoding a small peptide can induce the initial phase of ATR in guinea pigs.


Subject(s)
Ixodes , Tick Bites , Animals , Guinea Pigs , Tick Bites/immunology , Ixodes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Vaccination/methods , Salivary Proteins and Peptides/immunology , Salivary Proteins and Peptides/genetics , Epitopes/immunology , Female , RNA, Messenger/immunology , RNA, Messenger/genetics , Nanoparticles/chemistry , Erythema/immunology , Erythema/etiology , mRNA Vaccines , Liposomes
16.
BMC Med ; 22(1): 384, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267060

ABSTRACT

BACKGROUND: Extending the dosing interval of a primary series of mRNA COVID-19 vaccination has been employed to reduce myocarditis risk in adolescents, but previous evaluation of impact on vaccine effectiveness (VE) is limited to risk after second dose. METHODS: We quantified the impact of the dosing interval based on case notifications and vaccination uptake in Hong Kong from January to April 2022, based on calendar-time proportional hazards models and matching approaches. RESULTS: We estimated that the hazard ratio (HR) and odds ratio (OR) of infections after the second dose for extended (28 days or more) versus regular (21-27 days) dosing intervals ranged from 0.86 to 0.99 from calendar-time proportional hazards models, and from 0.85 to 0.87 from matching approaches, respectively. Adolescents in the extended dosing groups (including those who did not receive a second dose in the study period) had a higher hazard of infection than those with a regular dosing interval during the intra-dose period (HR 1.66; 95% CI 1.07, 2.59; p = 0.02) after the first dose. CONCLUSIONS: Implementing an extended dosing interval should consider multiple factors including the degree of myocarditis risk, the degree of protection afforded by each dose, and the extra protection achievable using an extended dosing interval.


Subject(s)
COVID-19 Vaccines , COVID-19 , Vaccine Efficacy , Humans , Adolescent , Male , COVID-19/prevention & control , COVID-19/epidemiology , Female , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Hong Kong/epidemiology , SARS-CoV-2/immunology , Immunization Schedule , Myocarditis/prevention & control , Myocarditis/epidemiology , Child , mRNA Vaccines , Proportional Hazards Models , Vaccination/methods
17.
Viruses ; 16(9)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39339880

ABSTRACT

Vaccination has led to significant dismantling of infectious diseases worldwide. Since the dawn of the SARS-CoV-2 pandemic, there has been increased popularity in the usage and study of the mRNA vaccine platform. Here, we highlight fundamental knowledge on mRNA vaccine pharmacology, followed by the immunity conferred by innate sensing and adaptive responses resulting from exposure to the mRNA vaccine construct and encapsulation materials. A better understanding of these immune mechanisms will shed light on further improvements in mRNA vaccine design, aiming to improve efficiency and optimize immune responses upon inoculation.


Subject(s)
Adaptive Immunity , COVID-19 Vaccines , COVID-19 , Immunity, Innate , SARS-CoV-2 , Vaccination , mRNA Vaccines , Humans , COVID-19 Vaccines/immunology , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , mRNA Vaccines/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Animals , RNA, Messenger/genetics , RNA, Messenger/immunology
18.
J Virol ; 98(9): e0057424, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194250

ABSTRACT

Boosting with mRNA vaccines encoding variant-matched spike proteins has been implemented to mitigate their reduced efficacy against emerging SARS-CoV-2 variants. Nonetheless, in humans, it remains unclear whether boosting in the ipsilateral or contralateral arm with respect to the priming doses impacts immunity and protection. Here, we boosted K18-hACE2 mice with either monovalent mRNA-1273 (Wuhan-1 spike) or bivalent mRNA-1273.214 (Wuhan-1 + BA.1 spike) vaccine in the ipsilateral or contralateral leg after a two-dose priming series with mRNA-1273. Boosting in the ipsilateral or contralateral leg elicited equivalent levels of serum IgG and neutralizing antibody responses against Wuhan-1 and BA.1. While contralateral boosting with mRNA vaccines resulted in the expansion of spike-specific B and T cells beyond the ipsilateral draining lymph node (DLN) to the contralateral DLN, administration of a third mRNA vaccine dose at either site resulted in similar levels of antigen-specific germinal center B cells, plasmablasts/plasma cells, T follicular helper cells, and CD8+ T cells in the DLNs and the spleen. Furthermore, ipsilateral and contralateral boosting with mRNA-1273 or mRNA-1273.214 vaccines conferred similar homologous or heterologous immune protection against SARS-CoV-2 BA.1 virus challenge with equivalent reductions in viral RNA and infectious virus in the nasal turbinates and lungs. Collectively, our data show limited differences in B and T cell immune responses after ipsilateral and contralateral site boosting by mRNA vaccines that do not substantively impact protection against an Omicron strain.IMPORTANCESequential boosting with mRNA vaccines has been an effective strategy to overcome waning immunity and neutralization escape by emerging SARS-CoV-2 variants. However, it remains unclear how the site of boosting relative to the primary vaccination series shapes optimal immune responses or breadth of protection against variants. In K18-hACE2 transgenic mice, we observed that intramuscular boosting with historical monovalent or variant-matched bivalent vaccines in the ipsilateral or contralateral limb elicited comparable levels of serum spike-specific antibody and antigen-specific B and T cell responses. Moreover, boosting on either side conferred equivalent protection against a SARS-CoV-2 Omicron challenge strain. Our data in mice suggest that the site of intramuscular boosting with an mRNA vaccine does not substantially impact immunity or protection against SARS-CoV-2 infection.


Subject(s)
2019-nCoV Vaccine mRNA-1273 , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , mRNA Vaccines , Animals , SARS-CoV-2/immunology , Mice , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , 2019-nCoV Vaccine mRNA-1273/immunology , mRNA Vaccines/immunology , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Humans , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism
19.
J Virol ; 98(9): e0053524, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39158273

ABSTRACT

Coronaviruses (CoVs) are important pathogens for humans and other vertebrates, causing severe respiratory and intestinal infections that have become a threat to public health because of the potential for interspecies transmission between animals and humans. Therefore, the development of safe, effective vaccines remains a top priority for the control of CoV infection. The unique immunological characteristics of vaccines featuring messenger RNA (mRNA) present an advantageous tool for coronavirus vaccine development. Here, we designed two lipid nanoparticle (LNP)-encapsulated mRNA (mRNA-LNP) vaccines: one encoding full-length spike (S) protein and the other encoding the spike ectodomain (Se) from porcine deltacoronavirus (PDCoV). Fourteen days after primary immunization, both mRNA vaccines induced high levels of immunoglobulin G and neutralizing antibodies in mice, with the S vaccine showing better performance than the Se vaccine. Passive immune protection of the S mRNA vaccine in suckling piglets was confirmed by the induction of robust PDCoV-specific humoral and cellular immune responses. The S mRNA vaccine also showed better protective effects than the inactivated vaccine. Our results suggest that the novel PDCoV-S mRNA-LNP vaccine may have the potential to combat PDCoV infection. IMPORTANCE: As an emerging porcine enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) has the potential for cross-species transmission, attracting extensive attention. Messenger RNA (mRNA) vaccines are a promising option for combating emerging and re-emerging infectious diseases, as evidenced by the demonstrated efficacy of the COVID-19 mRNA vaccine. Here, we first demonstrated that PDCoV-S mRNA-lipid nanoparticle (LNP) vaccines could induce potent humoral and cellular immune responses in mice. An evaluation of passive immune protection of S mRNA vaccines in suckling piglets confirmed that the protective effect of mRNA vaccine was better than that of inactivated vaccine. This study suggests that the PDCoV-S mRNA-LNP vaccine may serve as a potential and novel vaccine candidate for combating PDCoV infection.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Coronavirus Infections , Spike Glycoprotein, Coronavirus , Swine Diseases , Viral Vaccines , Animals , Swine , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Mice , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , mRNA Vaccines , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Nanoparticles , RNA, Messenger/genetics , RNA, Messenger/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice, Inbred BALB C , Female , Immunity, Humoral , Liposomes
20.
Nat Commun ; 15(1): 7358, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39191745

ABSTRACT

Whole virion inactivated vaccine CoronaVac (C) and Spike (S) mRNA BNT162b2 (B) vaccines differ greatly in their ability to elicit neutralizing antibodies but have somewhat comparable effectiveness in protecting from severe COVID-19. We conducted further analyses for a randomized trial (Cobovax study, NCT05057169) of third dose homologous and heterologous booster vaccination, i.e. four interventions CC-C, CC-B, BB-C and BB-B. Here, we assess vaccine immunogenicity beyond neutralizing function, including S and non-S antibodies with Fc receptor (FcR) binding, antibody avidity and T cell specificity to 6 months post-vaccination. Ancestral and Omicron S-specific IgG and FcR binding are significantly higher by BNT162b2 booster than CoronaVac, regardless of first doses. Nucleocapsid (N) antibodies are only increased in homologous boosted CoronaVac participants (CC-C). CoronaVac primed participants have lower baseline S-specific CD4+ IFNγ+ cells, but are significantly increased by either CoronaVac or BNT162b2 boosters. Priming vaccine content defined T cell peptide specificity preference, with S-specific T cells dominating B primed groups and non-S structural peptides contributing more in C primed groups, regardless of booster type. S-specific CD4+ T cell responses, N-specific antibodies, and antibody effector functions via Fc receptor binding may contribute to protection and compensate for less potent neutralizing responses in CoronaVac recipients.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19 Vaccines , COVID-19 , Receptors, Fc , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccines, Inactivated , mRNA Vaccines , Humans , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , Receptors, Fc/immunology , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , BNT162 Vaccine/immunology , BNT162 Vaccine/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , mRNA Vaccines/immunology , Spike Glycoprotein, Coronavirus/immunology , Female , Immunization, Secondary , Immunogenicity, Vaccine , Adult , T-Lymphocytes/immunology , Male , Immunoglobulin G/immunology , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL