Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.995
Filter
2.
J Nanobiotechnology ; 22(1): 295, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807131

ABSTRACT

The signal sequence played a crucial role in the efficacy of mRNA vaccines against virus pandemic by influencing antigen translation. However, limited research had been conducted to compare and analyze the specific mechanisms involved. In this study, a novel approach was introduced by substituting the signal sequence of the mRNA antigen to enhance its immune response. Computational simulations demonstrated that various signal peptides differed in their binding capacities with the signal recognition particle (SRP) 54 M subunit, which positively correlated with antigen translation efficiency. Our data revealed that the signal sequences of tPA and IL-6-modified receptor binding domain (RBD) mRNA vaccines sequentially led to higher antigen expression and elicited more robust humoral and cellular immune protection against the SARS-CoV-2 compared to the original signal sequence. By highlighting the importance of the signal sequence, this research provided a foundational and safe approach for ongoing modifications in signal sequence-antigen design, aiming to optimize the efficacy of mRNA vaccines.


Subject(s)
Protein Sorting Signals , SARS-CoV-2 , mRNA Vaccines , Animals , Mice , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19/immunology , Mice, Inbred BALB C , RNA, Messenger/genetics , COVID-19 Vaccines/immunology , Female , Humans , Antigens, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/chemistry , Antibodies, Viral/immunology , Immunity, Humoral , Vaccines, Synthetic/immunology , Immunity, Cellular
3.
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
4.
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)
Antibodies, Viral , Cross Reactions , Immunity, Humoral , Influenza Vaccines , mRNA Vaccines , Influenza Vaccines/immunology , Animals , mRNA Vaccines/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Humans , Cross Reactions/immunology , Mice , Influenza, Human/prevention & control , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Female , Seasons , Immunogenicity, Vaccine , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Mice, Inbred BALB C , Influenza A Virus, H1N1 Subtype/immunology , COVID-19/prevention & control , COVID-19/immunology , Vaccination
5.
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
6.
J Med Virol ; 96(6): e29710, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38804187

ABSTRACT

Kidney transplant recipients (KTRs), like other solid organ transplant recipients display a suboptimal response to mRNA vaccines, with only about half achieving seroconversion after two doses. However, the effectiveness of a booster dose, particularly in generating neutralizing antibodies (NAbs), remains poorly understood, as most studies have mainly focused on non-neutralizing antibodies. Here, we have longitudinally assessed the humoral response to the SARS-CoV-2 mRNA vaccine in 40 KTRs over a year, examining changes in both anti-spike IgG and NAbs following a booster dose administered about 5 months post-second dose. We found a significant humoral response increase 5 months post-booster, a stark contrast to the attenuated response observed after the second dose. Of note, nearly a quarter of participants did not achieve protective plasma levels even after the booster dose. We also found that the higher estimated glomerular filtration rate (eGFR) correlated with a more robust humoral response postvaccination. Altogether, these findings underscore the effectiveness of the booster dose in enhancing durable humoral immunity in KTRs, as evidenced by the protective level of NAbs found in 65% of the patients 5 months post- booster, especially those with higher eGFR rates.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunity, Humoral , Immunization, Secondary , Kidney Transplantation , SARS-CoV-2 , Transplant Recipients , Humans , Kidney Transplantation/adverse effects , Male , Antibodies, Viral/blood , Female , Middle Aged , Antibodies, Neutralizing/blood , COVID-19/prevention & control , COVID-19/immunology , Prospective Studies , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Aged , Adult , Immunoglobulin G/blood , Monitoring, Immunologic/methods , mRNA Vaccines , Spike Glycoprotein, Coronavirus/immunology , Longitudinal Studies
7.
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
8.
Expert Rev Vaccines ; 23(1): 570-583, 2024.
Article in English | MEDLINE | ID: mdl-38733272

ABSTRACT

INTRODUCTION: The mRNA vaccine technologies have progressed rapidly in recent years. The COVID-19 pandemic has accelerated the application of mRNA vaccines, with research and development and clinical trials underway for many vaccines. Application of the quality by design (QbD) framework to mRNA vaccine development and establishing standardized quality control protocols for mRNA vaccines are essential for the continued development of high-quality mRNA vaccines. AREAS COVERED: mRNA vaccines include linear mRNA, self-amplifying mRNA, and circular RNA vaccines. This article summarizes the progress of research on quality control of these three types of vaccines and presents associated challenges and considerations. EXPERT OPINION: Although there has been rapid progress in research on linear mRNA vaccines, their degradation patterns remain unclear. In addition, standardized assays for key impurities, such as residual dsRNA and T7 RNA polymerase, are still lacking. For self-amplifying mRNA vaccines, a key focus should be control of stability in vivo and in vitro. For circular RNA vaccines, standardized assays, and reference standards for determining degree of circularization should be established and optimized.


Subject(s)
COVID-19 Vaccines , COVID-19 , Quality Control , mRNA Vaccines , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/standards , COVID-19/prevention & control , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccine Development , Animals , RNA, Messenger/genetics , RNA, Messenger/immunology , SARS-CoV-2/immunology , SARS-CoV-2/genetics
9.
Hum Vaccin Immunother ; 20(1): 2350091, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38757631

ABSTRACT

Although previous studies have shown no increased mortality risk after the primary series of COVID-19 mRNA vaccines, reports on booster doses are lacking. This study aimed to evaluate mortality risk after the mRNA vaccine boosters in addition to the primary series. This nested case-control study included two age-specific cohorts (18-64 and ≥65 years as of February 1, 2021) in two municipalities. All deaths were identified and matched five controls for each case at each date of death (index date) with risk set sampling according to municipality, age, and sex. The adjusted odds ratios (aORs) and 95% confidence intervals (CIs) for mRNA vaccines (first to fifth doses) were estimated by comparing with no vaccination within 21 and 42 days before the index date using a conditional logistic regression model. The 18-64-years cohort comprised 431 cases (mean age, 57.0 years; men, 58.2%) and 2,155 controls (mean age, 56.0; men, 58.2%), whereas the ≥65-years cohort comprised 12,166 cases (84.0; 50.2%) and 60,830 controls (84.0, 50.2%). The aORs (95% CI) in 0-21 days after the third and fourth doses in the 18-64-years cohort were 0.62 (0.24, 1.62) and 0.38 (0.08, 1.84), respectively. The aORs (95% CI) after the third to fifth doses in the ≥65 years cohort were 0.36 (0.31, 0.43), 0.30 (0.25, 0.37), and 0.26 (0.20, 0.33), respectively. In conclusion, booster doses of mRNA vaccines do not increase mortality risk. These findings could help subsequent vaccine campaigns and alleviate vaccine hesitancy.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , mRNA Vaccines , Humans , Male , Middle Aged , Female , COVID-19/prevention & control , COVID-19/mortality , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Adult , Aged , Case-Control Studies , Young Adult , Japan/epidemiology , Adolescent , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccination/statistics & numerical data
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.
Int Immunopharmacol ; 134: 112192, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38761778

ABSTRACT

The recurrent COVID-19 infection, despite global vaccination, highlights the need for booster doses. A heterologous booster has been suggested to enhance immunity and protection against emerging variants of concern of the SARS-CoV-2 virus. In this report, we aimed to assess the safety, and immunogenicity of COReNAPCIN, as a fourth booster dose after three doses of inactivated vaccines. METHODS: The study was conducted as a double-blind, randomized, placebo-controlled phase 1 clinical trial of the mRNA-based vaccine candidate, COReNAPCIN. The vaccine was injected as a heterologous booster in healthy Iranian adults aged 18-50 who had previously received three doses of inactivated SARS-CoV-2 vaccines. In the study, 30 participants were randomly assigned to receive either COReNAPCIN in two different doses (25 µg and 50 µg) or placebo. The vaccine candidate contained mRNA encoding the complete sequence of the pre-fusion stabilized Spike protein of SARS-CoV-2, formulated within lipid nanoparticles. The primary endpoint was safety and the secondary objective was humoral immunogenicity until 6 months post-vaccination. The cellular immunogenicity was pursued as an exploratory outcome. RESULTS: COReNAPCIN was well tolerated in vaccinated individuals in both doses with no life-threatening or other serious adverse events. The most noticeable solicited adverse events were pain at the site of injection, fatigue and myalgia. Regarding the immunogenicity, despite the seroprevalence of SARS-CoV-2 antibodies due to the vaccination history for all and previous SARS-CoV-2 infection for some participants, the recipients of 25 and 50 µg COReNAPCIN, two weeks post-vaccination, showed 6·6 and 8·1 fold increase in the level of anti-RBD, and 11·5 and 21·7 fold increase in the level of anti-spike antibody, respectively. The geometric mean virus neutralizing titers reached 10.2 fold in the 25 µg group and 8.4 fold in 50 µg group of pre-boost levels. After 6 months, the measured anti-spike antibody concentration still maintains a geometric mean fold rise of 2.8 and 6.3, comparing the baseline levels in 25 and 50 µg groups, respectively. Additionally, the significant increase in the spike-specific IFN-ϒ T-cell response upon vaccination underscores the activation of cellular immunity. CONCLUSION: COReNAPCIN booster showed favorable safety, tolerability, and immunogenicity profile, supporting its further clinical development (Trial registration: IRCT20230131057293N1).


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , Humans , Adult , Male , Double-Blind Method , Female , COVID-19 Vaccines/immunology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/administration & dosage , Iran , SARS-CoV-2/immunology , Young Adult , COVID-19/prevention & control , COVID-19/immunology , Middle Aged , Antibodies, Viral/blood , Immunogenicity, Vaccine , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Neutralizing/blood , mRNA Vaccines , Adolescent , Follow-Up Studies
12.
Methods Mol Biol ; 2786: 1-22, 2024.
Article in English | MEDLINE | ID: mdl-38814388

ABSTRACT

Available prophylactic vaccines help prevent many infectious diseases that burden humanity. Future vaccinology will likely extend these benefits by more effectively countering newly emerging pathogens, fighting currently intractable infections, or even generating novel treatment modalities for non-infectious diseases. Instead of applying protein antigen directly, RNA vaccines contain short-lived genetic information that guides the expression of protein antigen in the vaccinee, like infection with a recombinant viral vector. Upon decades of research, messenger RNA-lipid nanoparticle (mRNA-LNP) vaccines have proven clinical value in addressing the COVID-19 pandemic as they combine benefits of killed subunit vaccines and live-attenuated vectors, including flexible production, self-adjuvanting effects, and stimulation of humoral and cellular immunity. RNA vaccines remain subject to continued development raising high hopes for broader future application. Their mechanistic versatility promises to make them a key tool of vaccinology and immunotherapy going forward. Here, I briefly review key developments in RNA vaccines and outline the contents of this volume of Methods in Molecular Biology.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , mRNA Vaccines , Humans , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Nanoparticles/chemistry , Lipids/chemistry , Vaccines, Synthetic/immunology , Liposomes
13.
Virol J ; 21(1): 124, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822328

ABSTRACT

Cervical cancer (CC) and other malignant malignancies are acknowledged to be primarily caused by persistent human papillomavirus (HPV) infection. Historically, vaccinations against viruses that produce neutralizing antibodies unique to the virus have been an affordable way to manage viral diseases. CC risk is decreased, but not eliminated, by HPV vaccinations. Since vaccinations have been made available globally, almost 90% of HPV infections have been successfully avoided. On the lesions and diseases that are already present, however, no discernible treatment benefit has been shown. As a result, therapeutic vaccines that elicit immune responses mediated by cells are necessary for the treatment of established infections and cancers. mRNA vaccines possess remarkable potential in combating viral diseases and malignancy as a result of their superior industrial production, safety, and efficacy. Furthermore, considering the expeditiousness of production, the mRNA vaccine exhibits promise as a therapeutic approach targeting HPV. Given that the HPV-encoded early proteins, including oncoproteins E6 and E7, are consistently present in HPV-related cancers and pre-cancerous lesions and have crucial functions in the progression and persistence of HPV-related diseases, they serve as ideal targets for therapeutic HPV vaccines. The action mechanism of HPV and HPV-related cancer mRNA vaccines, their recent advancements in clinical trials, and the potential for their therapeutic applications are highlighted in this study, which also offers a quick summary of the present state of mRNA vaccines. Lastly, we highlight a few difficulties with mRNA HPV vaccination clinical practice and provide our thoughts on further advancements in this quickly changing sector. It is expected that mRNA vaccines will soon be produced quickly for clinical HPV prevention and treatment.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , mRNA Vaccines , Humans , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Infections/immunology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/therapy , Female , Papillomaviridae/immunology , Papillomaviridae/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Human Papillomavirus Viruses
14.
Med Sci (Basel) ; 12(2)2024 May 22.
Article in English | MEDLINE | ID: mdl-38804384

ABSTRACT

mRNA vaccines have emerged as an optimistic technological platform for vaccine innovation in this new scientific era. mRNA vaccines have dramatically altered the domain of vaccinology by offering a versatile and rapid approach to combating infectious diseases and virus-induced cancers. Clinical trials have demonstrated efficacy rates of 94-95% in preventing COVID-19, and mRNA vaccines have been increasingly recognized as a powerful vaccine platform. Although mRNA vaccines have played an essential role in the COVID-19 pandemic, they still have several limitations; their instability and degradation affect their storage, delivery, and over-all efficiency. mRNA is typically enclosed in a transport mechanism to facilitate its entry into the target cell because it is an unstable and negatively charged molecule. For instance, mRNA that is given using lipid-nanoparticle-based vaccine delivery systems (LNPs) solely enters cells through endocytosis, establishing an endosome without damaging the cell membrane. The COVID-19 pandemic has accelerated the development of mRNA vaccine platforms used to treat and prevent several infectious diseases. This technology has the potential to change the future course of the disease by providing a safe and effective way to combat infectious diseases and cancer. A single-stranded genetic sequence found in mRNA vaccines instructs host cells to produce proteins inside ribosomes to elicit immunological responses and prepare the immune system to fight infections or cancer cells. The potential applications of mRNA vaccine technology are vast and can lead to the development of a preferred vaccine pattern. As a result, a new generation of vaccinations has gradually gained popularity and access to the general population. To adapt the design of an antigen, and even combine sequences from different variations in response to new changes in the viral genome, mRNA vaccines may be used. Current mRNA vaccines provide adequate safety and protection, but the duration of that protection can only be determined if further clinical research is conducted.


Subject(s)
COVID-19 , SARS-CoV-2 , mRNA Vaccines , Humans , COVID-19/prevention & control , SARS-CoV-2/immunology , Pandemics/prevention & control , Oncogenic Viruses , Vaccines, Synthetic , Vaccine Development , COVID-19 Vaccines/immunology , Pneumonia, Viral/prevention & control , Coronavirus Infections/prevention & control , Betacoronavirus , Viral Vaccines/immunology , RNA, Messenger , Neoplasms
15.
Hum Vaccin Immunother ; 20(1): 2355037, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38813652

ABSTRACT

BACKGROUND: In recent years, infectious diseases like COVID-19 have had profound global socio-economic impacts. mRNA vaccines have gained prominence due to their rapid development, industrial adaptability, simplicity, and responsiveness to new variants. Notably, the 2023 Nobel Prize in Physiology or Medicine recognized significant contributions to mRNA vaccine research. METHODS: Our study employed a comprehensive bibliometric analysis using the Web of Science Core Collection (WoSCC) database, encompassing 5,512 papers on mRNA vaccines from 2003 to 2023. We generated cooperation maps, co-citation analyses, and keyword clustering to evaluate the field's developmental history and achievements. RESULTS: The analysis yielded knowledge maps highlighting countries/institutions, influential authors, frequently published and highly cited journals, and seminal references. Ongoing research hotspots encompass immune responses, stability enhancement, applications in cancer prevention and treatment, and combating infectious diseases using mRNA technology. CONCLUSIONS: mRNA vaccines represent a transformative development in infectious disease prevention. This study provides insights into the field's growth and identifies key research priorities, facilitating advancements in vaccine technology and addressing future challenges.


Subject(s)
Bibliometrics , COVID-19 , mRNA Vaccines , Humans , COVID-19/prevention & control , COVID-19/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Biomedical Research/trends , Vaccine Development , SARS-CoV-2/immunology , SARS-CoV-2/genetics , RNA, Messenger/genetics
16.
Methods Mol Biol ; 2786: 365-386, 2024.
Article in English | MEDLINE | ID: mdl-38814404

ABSTRACT

In this chapter, we will first consider the overall goal of nonclinical safety testing during drug development and have a brief overview of its regulatory background. We will then discuss some basic requirements of safety/toxicity testing before concentrating on the safety testing of RNA vaccines and developing a sample RNA vaccine safety testing program.


Subject(s)
mRNA Vaccines , Animals , Humans , Drug Evaluation, Preclinical/methods , Toxicity Tests/methods
17.
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
18.
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
19.
J Transl Med ; 22(1): 432, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715088

ABSTRACT

BACKGROUND: SARS-CoV-2 mRNA vaccines are highly immunogenic in people living with HIV (PLWH) on effective antiretroviral therapy (ART). However, whether viro-immunologic parameters or other factors affect immune responses to vaccination is debated. This study aimed to develop a machine learning-based model able to predict the humoral response to mRNA vaccines in PLWH and to assess the impact of demographic and clinical variables on antibody production over time. METHODS: Different machine learning algorithms have been compared in the setting of a longitudinal observational study involving 497 PLWH, after primary and booster SARS-CoV-2 mRNA vaccination. Both Generalized Linear Models and non-linear Models (Tree Regression and Random Forest) were trained and tested. RESULTS: Non-linear algorithms showed better ability to predict vaccine-elicited humoral responses. The best-performing Random Forest model identified a few variables as more influential, within 39 clinical, demographic, and immunological factors. In particular, previous SARS-CoV-2 infection, BMI, CD4 T-cell count and CD4/CD8 ratio were positively associated with the primary cycle immunogenicity, yet their predictive value diminished with the administration of booster doses. CONCLUSIONS: In the present work we have built a non-linear Random Forest model capable of accurately predicting humoral responses to SARS-CoV-2 mRNA vaccination, and identifying relevant factors that influence the vaccine response in PLWH. In clinical contexts, the application of this model provides promising opportunities for predicting individual vaccine responses, thus facilitating the development of vaccination strategies tailored for PLWH.


Subject(s)
COVID-19 Vaccines , COVID-19 , HIV Infections , Immunity, Humoral , Immunization, Secondary , Machine Learning , SARS-CoV-2 , Humans , Male , Female , HIV Infections/immunology , Middle Aged , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Vaccination , Adult , Antibodies, Viral/blood , Antibodies, Viral/immunology , mRNA Vaccines , Longitudinal Studies , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Hum Vaccin Immunother ; 20(1): 2342592, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38714327

ABSTRACT

Messenger ribonucleic acid (mRNA) technology has been rapidly applied for the development of the COVID-19 vaccine. However, naked mRNA itself is inherently unstable. Lipid nanoparticles (LNPs) protect mRNAs from extracellular ribonucleases and facilitate mRNA trafficking. For mRNA vaccines, antigen-presenting cells utilize LNPs through uptake to elicit antigen-specific immunity. There are reports on the impact of various physical characteristics of LNPs, particularly those with sizes less than 200 nm, especially 50 to 150 nm, on the overall stability and protective efficacy of mRNA vaccines. To address this, a single change in the size of LNPs using the same mRNA stock solution was assessed for the physicochemical characterization of the resulting mRNA-LNPs vaccine, along with the evaluation of their protective efficacy. Particles of smaller sizes generally disperse more effectively in solutions, with minimized occurrence of particle precipitation and aggregation. Here, we demonstrate that the vaccine containing 80-100 nm mRNA-LNPs showed the best stability and protection at 4°C and -20°C. Furthermore, we can conclude that freezing the vaccine at -20°C is more appropriate for maintaining stability over the long term. This effort is poised to provide a scientific basis for improving the quality of ongoing mRNA vaccine endeavors and providing information on the development of novel products.


Subject(s)
COVID-19 Vaccines , COVID-19 , Lipids , Nanoparticles , Particle Size , SARS-CoV-2 , mRNA Vaccines , Nanoparticles/chemistry , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , Lipids/chemistry , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Animals , Mice , Antibodies, Viral/immunology , Female , RNA, Messenger/immunology , RNA, Messenger/genetics , Drug Stability , Immunogenicity, Vaccine , Humans , Mice, Inbred BALB C , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Liposomes
SELECTION OF CITATIONS
SEARCH DETAIL
...