Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.060
Filter
1.
Oncoimmunology ; 13(1): 2373526, 2024.
Article in English | MEDLINE | ID: mdl-38948931

ABSTRACT

Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.


Subject(s)
Adjuvants, Immunologic , Antigens, Neoplasm , Cancer Vaccines , Dendritic Cells , Prostatic Neoplasms , RNA, Messenger , Animals , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/drug therapy , Antigens, Neoplasm/immunology , Mice , Dendritic Cells/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Humans , Mice, Inbred C57BL , Cell Line, Tumor , mRNA Vaccines , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Immunotherapy/methods , Lymphocyte Activation/drug effects
2.
PLoS One ; 19(7): e0305413, 2024.
Article in English | MEDLINE | ID: mdl-38976715

ABSTRACT

Pancreatic ductal adenocarcinoma is the most prevalent pancreatic cancer, which is considered a significant global health concern. Chemotherapy and surgery are the mainstays of current pancreatic cancer treatments; however, a few cases are suitable for surgery, and most of the cases will experience recurrent episodes. Compared to DNA or peptide vaccines, mRNA vaccines for pancreatic cancer have more promise because of their delivery, enhanced immune responses, and lower proneness to mutation. We constructed an mRNA vaccine by analyzing S100 family proteins, which are all major activators of receptors for advanced glycation end products. We applied immunoinformatic approaches, including physicochemical properties analysis, structural prediction and validation, molecular docking study, in silico cloning, and immune simulations. The designed mRNA vaccine was estimated to have a molecular weight of 165023.50 Da and was highly soluble (grand average of hydropathicity of -0.440). In the structural assessment, the vaccine seemed to be a well-stable and functioning protein (Z score of -8.94). Also, the docking analysis suggested that the vaccine had a high affinity for TLR-2 and TLR-4 receptors. Additionally, the molecular mechanics with generalized Born and surface area solvation analysis of the "Vaccine-TLR-2" (-141.07 kcal/mol) and "Vaccine-TLR-4" (-271.72 kcal/mol) complexes also suggests a strong binding affinity for the receptors. Codon optimization also provided a high expression level with a GC content of 47.04% and a codon adaptation index score 1.0. The appearance of memory B-cells and T-cells was also observed over a while, with an increased level of helper T-cells and immunoglobulins (IgM and IgG). Moreover, the minimum free energy of the mRNA vaccine was predicted at -1760.00 kcal/mol, indicating the stability of the vaccine following its entry, transcription, and expression. This hypothetical vaccine offers a groundbreaking tool for future research and therapeutic development of pancreatic cancer.


Subject(s)
Cancer Vaccines , Molecular Docking Simulation , Pancreatic Neoplasms , Pancreatic Neoplasms/immunology , Humans , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , mRNA Vaccines/immunology , Computational Biology/methods , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Vaccinology/methods , Toll-Like Receptor 2/immunology , Computer Simulation , RNA, Messenger/genetics , RNA, Messenger/immunology , Immunoinformatics
3.
Emerg Microbes Infect ; 13(1): 2377606, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38979723

ABSTRACT

The lack of success in clinical trials for HIV vaccines highlights the need to explore novel strategies for vaccine development. Research on highly exposed seronegative (HESN) HIV-resistant Kenyan female sex workers revealed naturally protective immunity is correlated with a focused immune response mediated by virus-specific CD8 T cells. Further studies indicated that the immune response is unconventionally focused on highly conserved sequences around HIV viral protease cleavage sites (VPCS). Thus, taking an unconventional approach to HIV vaccine development, we designed lipid nanoparticles loaded with mRNA that encodes multi-epitopes of VPCS (MEVPCS-mRNA LNP), a strategic design to boost antigen presentation by dendritic cells, promoting effective cellular immunity. Furthermore, we developed a novel cold-chain compatible mRNA LNP formulation, ensuring long-term stability and compatibility with cold-chain storage/transport, widening accessibility of mRNA LNP vaccine in low-income countries. The in-vivo mouse study demonstrated that the vaccinated group generated VPCS-specific CD8 memory T cells, both systemically and at mucosal sites of viral entry. The MEVPCS-mRNA LNP vaccine-induced CD8 T cell immunity closely resembled that of the HESN group and displayed a polyfunctional profile. Notably, it induced minimal to no activation of CD4 T cells. This proof-of-concept study underscores the potential of the MEVPCS-mRNA LNP vaccine in eliciting CD8 T cell memory specific to the highly conserved multiple VPCS, consequently having a broad coverage in human populations and limiting viral escape mutation. The MEVPCS-mRNA LNP vaccine holds promise as a candidate for an effective prophylactic HIV vaccine.


Subject(s)
AIDS Vaccines , CD8-Positive T-Lymphocytes , HIV Infections , mRNA Vaccines , AIDS Vaccines/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Female , HIV Infections/prevention & control , HIV Infections/immunology , HIV Infections/virology , Humans , HIV-1/immunology , HIV-1/genetics , Nanoparticles/chemistry , HIV Protease/genetics , HIV Protease/immunology , Kenya , Sex Workers , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes/immunology , Epitopes/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Liposomes
4.
Nat Commun ; 15(1): 6181, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39039113

ABSTRACT

The long-term association between mRNA-based coronavirus disease 2019 (COVID-19) vaccination and the development of autoimmune connective tissue diseases (AI-CTDs) remains unclear. In this nationwide, population-based cohort study involving 9,258,803 individuals, we aim to determine whether the incidence of AI-CTDs is associated with mRNA vaccination. The study spans over 1 year of observation and further analyses the risk of AI-CTDs by stratifying demographics and vaccination profiles and treating booster vaccination as time-varying covariate. We report that the risk of developing most AI-CTDs did not increase following mRNA vaccination, except for systemic lupus erythematosus with a 1.16-fold risk in vaccinated individuals relative to controls. Comparable results were reported in the stratified analyses for age, sex, mRNA vaccine type, and prior history of non-mRNA vaccination. However, a booster vaccination was associated with an increased risk of some AI-CTDs including alopecia areata, psoriasis, and rheumatoid arthritis. Overall, we conclude that mRNA-based vaccinations are not associated with an increased risk of most AI-CTDs, although further research is needed regarding its potential association with certain conditions.


Subject(s)
Autoimmune Diseases , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccination , Humans , Autoimmune Diseases/epidemiology , Autoimmune Diseases/genetics , Female , Male , Middle Aged , Adult , Republic of Korea/epidemiology , COVID-19/prevention & control , COVID-19/epidemiology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Cohort Studies , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Aged , Young Adult , Incidence , Adolescent , Connective Tissue Diseases/genetics , Connective Tissue Diseases/epidemiology , mRNA Vaccines , Immunization, Secondary
5.
Medicine (Baltimore) ; 103(27): e38666, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968513

ABSTRACT

Adenocarcinoma of the pancreas (PAAD) is one of the deadliest malignant tumors, and messenger ribonucleic acid vaccines, which constitute the latest generation of vaccine technology, are expected to lead to new ideas for the treatment of pancreatic cancer. The Cancer Genome Atlas-PAAD and Genotype-Tissue Expression data were merged and analyzed. Weighted gene coexpression network analysis was used to identify gene modules associated with tumor mutational burden among the genes related to both immunity and oxidative stress. Differentially expressed immune-related oxidative stress genes were screened via univariate Cox regression analysis, and these genes were analyzed via nonnegative matrix factorization. After immune infiltration analysis, least absolute shrinkage and selection operator regression combined with Cox regression was used to construct the model, and the usefulness of the model was predicted based on the receiver operating characteristic curve and decision curve analysis curves after model construction. Finally, metabolic pathway enrichment was analyzed using gene set enrichment analysis combined with Kyoto Encyclopedia of Genes and Genomes and gene ontology biological process analyses. This model consisting of the ERAP2, mesenchymal-epithelial transition factor (MET), CXCL9, and angiotensinogen (AGT) genes can be used to help predict the prognosis of pancreatic cancer patients more accurately than existing models. ERAP2 is involved in immune activation and is important in cancer immune evasion. MET binds to hepatocyte growth factor, leading to the dimerization and phosphorylation of c-MET. This activates various signaling pathways, including MAPK and PI3K, to regulate the proliferation, invasion, and migration of cancer cells. CXCL9 overexpression is associated with a poor patient prognosis and reduces the number of CD8 + cytotoxic T lymphocytes in the PAAD tumor microenvironment. AGT is cleaved by the renin enzyme to produce angiotensin 1, and AGT-converting enzyme cleaves angiotensin 1 to produce angiotensin 2. Exposure to AGT-converting enzyme inhibitors after pancreatic cancer diagnosis is associated with improved survival. The 4 genes identified in the present study - ERAP2, MET, CXCL9, and AGT - are expected to serve as targets for messenger ribonucleic acid vaccine development and need to be further investigated in depth.


Subject(s)
Oxidative Stress , Pancreatic Neoplasms , mRNA Vaccines , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Humans , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Angiotensinogen/genetics , Gene Expression Regulation, Neoplastic , Prognosis
6.
Int J Oncol ; 65(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38994758

ABSTRACT

Cancer is characterized by unlimited proliferation and metastasis, and traditional therapeutic strategies usually result in the acquisition of drug resistance, thus highlighting the need for more personalized treatment. mRNA vaccines transfer the gene sequences of exogenous target antigens into human cells through transcription and translation to stimulate the body to produce specific immune responses against the encoded proteins, so as to enable the body to obtain immune protection against said antigens; this approach may be adopted for personalized cancer therapy. Since the recent coronavirus pandemic, the development of mRNA vaccines has seen substantial progress and widespread adoption. In the present review, the development of mRNA vaccines, their mechanisms of action, factors influencing their function and the current clinical applications of the vaccine are discussed. A focus is placed on the application of mRNA vaccines in cancer, with the aim of highlighting unique advances and the remaining challenges of this novel and promising therapeutic approach.


Subject(s)
Cancer Vaccines , Neoplasms , Vaccine Development , mRNA Vaccines , Humans , Neoplasms/immunology , Neoplasms/therapy , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , COVID-19/prevention & control , COVID-19/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Precision Medicine/methods , Immunotherapy/methods
7.
J Am Chem Soc ; 146(28): 19218-19228, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38955767

ABSTRACT

The messenger RNA (mRNA) vaccines hold great significance in contagion prevention and cancer immunotherapy. However, safely and effectively harnessing innate immunity to stimulate robust and durable adaptive immune protection is crucial, yet challenging. In this study, we synthesized a library of stimuli-responsive bivalent ionizable lipids (srBiv iLPs) with smart molecular blocks responsive to esterase, H2O2, cytochrome P450, alkaline phosphatase, nitroreductase, or glutathione (GSH), aiming to leverage physiological cues to trigger fast lipid degradation, promote mRNA translation, and induce robust antitumor immunity via reactive oxygen species (ROS)-mediated boosting. After subcutaneous immunization, esterase-responsive vaccine (eBiv-mVac) was rapidly internalized and transported into the draining lymph nodes. It then underwent fast decaging and self-immolative degradation in esterase-rich antigen-presenting cells, releasing sufficient mRNA for antigen translation and massive reactive quinone methides to elevate ROS levels. This resulted in broad activation of innate immunity to boost T cell response, prompting a large number of primed antigen-specific CD8+ T cells to circulate and infiltrate into tumors (>1000-fold versus unvaccinated control), thereby orchestrating innate and adaptive immunity to control tumor growth. Moreover, by further combining our vaccination strategy with immune checkpoint blockade, we demonstrated a synergism that significantly amplified the magnitude and function of antigen-specific CD8+ T cells. This, in turn, caused potent systemic antitumor efficacy and prolonged survival with high complete response rate in xenograft and metastasis models. Overall, our generalized stimuli-responsive mRNA delivery platform promises a paradigm shift in the design of potent vaccines for cancer immunotherapy, as well as effective and precise carriers for gene editing, protein replacement, and cell engineering.


Subject(s)
CD8-Positive T-Lymphocytes , Immunity, Innate , Reactive Oxygen Species , Reactive Oxygen Species/metabolism , Immunity, Innate/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , mRNA Vaccines/chemistry , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , RNA, Messenger/immunology , RNA, Messenger/genetics , Humans , Mice, Inbred C57BL
8.
J Med Virol ; 96(7): e29793, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39023111

ABSTRACT

Various vaccine platforms were developed and deployed against the COVID-19 disease. The Fc-mediated functions of IgG antibodies are essential in the adaptive immune response elicited by vaccines. However, the long-term changes of protein subunit vaccines and their combinations with messenger RNA (mRNA) vaccines are unknown. A total of 272 serum and plasma samples were collected from individuals who received first to third doses of the protein subunit Medigen, the mRNA (BNT, Moderna), or the adenovector AstraZeneca vaccines. The IgG subclass level was measured using enzyme-linked immunosorbent assay, and Fc-N glycosylation was measured using liquid chromatography coupled to tandem mass spectrometry. Antibody-dependent-cellular-phagocytosis (ADCP) and complement deposition (ADCD) of anti-spike (S) IgG antibodies were measured by flow cytometry. IgG1 and 3 reached the highest anti-S IgG subclass level. IgG1, 2, and 4 subclass levels significantly increased in mRNA- and Medigen-vaccinated individuals. Fc-glycosylation was stable, except in female BNT vaccinees, who showed increased bisection and decreased galactosylation. Female BNT vaccinees had a higher anti-S IgG titer than that of males. ADCP declined in all groups. ADCD was significantly lower in AstraZeneca-vaccinated individuals. Each vaccine produced specific long-term changes in Fc structure and function. This finding is critical when selecting a vaccine platform or combination to achieve the desired immune response.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunoglobulin G , SARS-CoV-2 , Vaccines, Subunit , mRNA Vaccines , Humans , Immunoglobulin G/blood , Female , Antibodies, Viral/blood , Male , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Adult , Middle Aged , COVID-19 Vaccines/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Vaccines, Subunit/administration & dosage , Glycosylation , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Aged , RNA, Messenger/genetics , Young Adult , Protein Subunit Vaccines
9.
Front Immunol ; 15: 1384442, 2024.
Article in English | MEDLINE | ID: mdl-38947333

ABSTRACT

The One Health approach, which integrates the health of humans, animals, plants, and ecosystems at various levels, is crucial for addressing interconnected health threats. This is complemented by the advent of mRNA vaccines, which have revolutionized disease prevention. They offer broad-spectrum effectiveness and can be rapidly customized to target specific pathogens. Their utility extends beyond human medicine, showing potential in veterinary practices to control diseases and reduce the risk of zoonotic transmissions. This review place mRNA vaccines and One Health in the context of tick-borne diseases. The potential of these vaccines to confer cross-species immunity is significant, potentially disrupting zoonotic disease transmission cycles and protecting the health of both humans and animals, while reducing tick populations, infestations and circulation of pathogens. The development and application of mRNA vaccines for tick and tick-borne pathogens represent a comprehensive strategy in global health, fostering a healthier ecosystem for all species in our interconnected world.


Subject(s)
One Health , Tick-Borne Diseases , Ticks , mRNA Vaccines , Animals , Humans , Tick-Borne Diseases/prevention & control , Tick-Borne Diseases/immunology , Tick-Borne Diseases/transmission , Ticks/microbiology , Ticks/immunology , Zoonoses/prevention & control , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccines, Synthetic/immunology
10.
Adv Pharmacol ; 100: 247-288, 2024.
Article in English | MEDLINE | ID: mdl-39034054

ABSTRACT

In the past decade, in vitro transcribed messenger RNAs (IVT-mRNAs) have emerged as promising therapeutic molecules. The clinical success of COVID-19 mRNA vaccines developed by Pfizer-BioNTech and Moderna, have demonstrated that IVT-mRNAs can be safely and successfully used in a clinical setting, and efforts are underway to develop IVT-mRNAs for therapeutic applications. Current applications of mRNA-based therapy have been focused on (1) mRNA vaccines for infectious diseases and cancer treatment; (2) protein replacement therapy; (3) gene editing therapy; and (4) cell-reprogramming therapies. Due to the recent clinical progress of cell-based immunotherapies, the last direction-the use of IVT-mRNAs as a therapeutic approach to program immune cells for the treatment of cancer has received extensive attention from the cancer immunotherapy field. Myeloid cells are important components of our immune system, and they play critical roles in mediating disease progression and regulating immunity against diseases. In this chapter, we discussed the progress of using IVT-mRNAs as a therapeutic approach to program myeloid cells against cancer and other immune-related diseases. Towards this direction, we first reviewed the pharmacology of IVT-mRNAs and the biology of myeloid cells as well as myeloid cell-targeting therapeutics. We then presented a few cases of current IVT-mRNA-based approaches to target and reprogram myeloid cells for disease treatment and discussed the advantages and limitations of these approaches. Finally, we presented our considerations in designing mRNA-based approaches to target myeloid cells for disease treatment.


Subject(s)
Immunotherapy , Myeloid Cells , Neoplasms , RNA, Messenger , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/genetics , Immunotherapy/methods , Myeloid Cells/metabolism , Myeloid Cells/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , COVID-19/immunology , COVID-19/therapy , mRNA Vaccines , COVID-19 Vaccines/immunology
11.
J Med Virol ; 96(7): e29790, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38994662

ABSTRACT

The effect of COVID-19 booster vaccination on SARS-CoV-2 T-cell mediated immune responses in elderly nursing home residents has not been explored in depth. Thirty-nine elderly nursing home residents (median age, 91 years) were included, all fully vaccinated with mRNA vaccines. The frequency of and the integrated mean fluorescence (iMFI) for peripheral blood SARS-CoV-2-Spike reactive IFN-γ-producing CD4+ or CD8+ T cells before and after the first (Pre-3D and Post-3D) and second (Pre-4D and Post-4D) vaccine booster doses was determined using flow cytometry for an intracellular staining method. 3D increased significantly (p = 0.01) the percentage of participants displaying detectable SARS-CoV-2-T-cell responses compared with pre-3D (97% vs. 74%). The magnitude of the increase was statistically significant for CD8+ T cells (p = 0.007) but not for CD4+ T cells (p = 0.77). A trend towards higher frequencies of peripheral blood SARS-CoV-2-CD8+ T cells was observed post-3D compared with pre-3D (p = 0.06). The percentage of participants with detectable SARS-S-CoV-2 CD4+ T-cell responses decreased post-4D (p = 0.035). Following 4D, a nonsignificant decrease in the frequencies of both T cell subsets was noticed (p = 0.94 for CD8+ T cells and p = 0.06 for CD4+ T cells). iMFI data mirrored that of T-cell frequencies. The kinetics of SARS-CoV-2 CD8+ and CD4+ T cells following receipt of 3D and 4D were comparable across SARS-CoV-2-experienced and -naïve participants and between individuals receiving a homologous or heterologous vaccine booster. 3D increased the percentage of elderly nursing home residents displaying detectable SARS-CoV-2 T-cell responses but had a marginal effect on T-cell frequencies. The impact of 4D on SARS-CoV-2 T-cell responses was negligible; whether this was due to suboptimal priming or rapid waning could not be ascertained.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , Nursing Homes , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Aged, 80 and over , Male , Spike Glycoprotein, Coronavirus/immunology , Female , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , COVID-19/prevention & control , CD4-Positive T-Lymphocytes/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , Aged , Interferon-gamma , mRNA Vaccines
12.
Sci Rep ; 14(1): 17308, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068230

ABSTRACT

The SARS-CoV-2 outbreak has provoked more than 6 million deaths worldwide. The scarcity of effective treatments and its virulence converted the vaccines into an essential tool to face it. The most used vaccines were the mRNA, adenovirus vector, and inactivated whole-virus. However, nowadays, infants aged < 6 months are not eligible for any vaccines against COVID-19, and their immunization relies on passive immunity. In this research, we investigated the humoral and cellular immune response generated on newborns of SARS-CoV-2 vaccinated mothers with mRNA or viral vector (VV) vaccine employing Fourier transformed infrared (FTIR) spectroscopy in saliva samples. For this purpose, saliva samples of newborns and their mothers were collected; the population was divided into two groups, VV and mRNA, which were subdivided into three subgroups: before pregnancy (BP), at the first (FTP) and second (STP) trimesters of pregnancy. The samples were analyzed using FTIR spectroscopy, and the bands associated with the humoral and cellular immune responses, such as IgG, IgA, and IFN-γ were analyzed. The integrated areas were calculated and compared to elucidate the quantity of those immunoglobins and the cytokine. Likewise, the correlation of the humoral and cellular immune response between the newborns and their mothers and the correlation between cellular and humoral immune response was also evaluated. The VV vaccine produced a significant humoral and cellular immune response in newborns and their mothers when they received it at the STP compared with the mRNA vaccine, evidencing statistical significance. However, no correlation was observed between newborns and their mothers when the vaccine was applied in this trimester of pregnancy. When administered BP, the mRNA vaccine generated more humoral immunity in newborns and their mothers. Nevertheless, compared with the VV vaccine, it only showed statistical significance in the mothers, highlighting that IgG showed a moderate positive correlation between the newborns and their mothers.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccination , Humans , Female , Spectroscopy, Fourier Transform Infrared/methods , Infant, Newborn , COVID-19/prevention & control , COVID-19/immunology , Pregnancy , Vaccination/methods , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , Adult , Mothers , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Viral/analysis , Immunity, Humoral , Saliva/immunology , Immunity, Cellular , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin A/immunology , Immunoglobulin A/analysis , Interferon-gamma/metabolism , mRNA Vaccines/immunology
13.
Viruses ; 16(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39066318

ABSTRACT

Marek's disease (MD), caused by the Marek's disease virus, is a lymphoproliferative disease in chickens that can be controlled by vaccination. However, the current vaccines can limit tumor growth and death but not virus replication and transmission. The present study aimed to evaluate host responses following intramuscular injection of an mRNA vaccine encoding gB and pp38 proteins of the MDV within the first 36 h. The vaccine was injected in low and high doses using prime and prime-boost strategies. The expression of type I and II interferons (IFNs), a panel of interferon-stimulated genes, and two key antiviral cytokines, IL-1ß and IL-2, were measured in spleen and lungs after vaccination. The transcriptional analysis of the above genes showed significant increases in the expression of MDA5, Myd88, IFN-α, IFN-ß, IFN-γ, IRF7, OAS, Mx1, and IL-2 in both the spleen and lungs within the first 36 h of immunization. Secondary immunization increased expression of all the above genes in the lungs. In contrast, only IFN-γ, MDA5, MyD88, Mx1, and OAS showed significant upregulation in the spleen after the secondary immunization. This study shows that two doses of the MDV mRNA vaccine encoding gB and pp38 antigens activate innate and adaptive responses and induce an antiviral state in chickens.


Subject(s)
Chickens , Cytokines , Herpesvirus 2, Gallid , Marek Disease Vaccines , Marek Disease , Animals , Chickens/immunology , Marek Disease/prevention & control , Marek Disease/immunology , Marek Disease/virology , Marek Disease Vaccines/immunology , Marek Disease Vaccines/administration & dosage , Marek Disease Vaccines/genetics , Cytokines/metabolism , Cytokines/immunology , Herpesvirus 2, Gallid/immunology , Herpesvirus 2, Gallid/genetics , Lung/virology , Lung/immunology , Spleen/immunology , Spleen/virology , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/virology , mRNA Vaccines/immunology , Vaccination , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics
14.
Int J Nanomedicine ; 19: 7201-7214, 2024.
Article in English | MEDLINE | ID: mdl-39050877

ABSTRACT

Introduction: The clinical success of mRNA vaccine during the COVID-19 pandemic has inspired emerging approaches to elevate mRNA vaccine immunogenicity. Among them, antigen fusion protein designs for improved immune cell targeting have been shown to augment humoral immunity against small antigen targets. Methods: This research demonstrates that SARS-CoV-2 receptor binding domain (RBD) fusion with a minimalistic peptide segment of complement component 3b (C3b, residues 727-767) ligand can improve mRNA vaccine immunogenicity through antigen targeting to complement receptor 1 (CR1). We affirm vaccines' antigenicity and targeting ability towards specific receptors through Western blot and immunofluorescence assay. Furthermore, mice immunization studies help the investigation of the antibody responses. Results: Using SARS-CoV-2 Omicron RBD antigen, we compare mRNA vaccine formulations expressing RBD fusion protein with mouse C3b peptide (RBD-mC3), RBD fusion protein with mouse Fc (RBD-Fc), and wild-type RBD. Our results confirm the proper antigenicity and normal functionality of RBD-mC3. Upon validating comparable antigen expression by the different vaccine formulations, receptor-targeting capability of the fusion antigens is further confirmed. In mouse immunization studies, we show that while both RBD-mC3 and RBD-Fc elevate vaccine immunogenicity, RBD-mC3 leads to more sustained RBD-specific titers over the RBD-Fc design, presumably due to reduced antigenic diversion by the minimalistic targeting ligand. Conclusion: The study demonstrates a novel C3b-based antigen design strategy for immune cell targeting and mRNA vaccine enhancement.


Subject(s)
COVID-19 Vaccines , SARS-CoV-2 , Animals , Mice , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/pharmacology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/chemistry , Immunogenicity, Vaccine , COVID-19/prevention & control , COVID-19/immunology , mRNA Vaccines , Female , Mice, Inbred BALB C , Humans , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry
15.
Nat Commun ; 15(1): 6421, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080316

ABSTRACT

The rodent-borne Andes virus (ANDV) causes a severe disease in humans. We developed an ANDV mRNA vaccine based on the M segment of the viral genome, either with regular uridine (U-mRNA) or N1-methylpseudouridine (m1Ψ-mRNA). Female mice immunized by m1Ψ-mRNA developed slightly greater germinal center (GC) responses than U-mRNA-immunized mice. Single cell RNA and BCR sequencing of the GC B cells revealed similar levels of activation, except an additional cluster of cells exhibiting interferon response in animals vaccinated with U-mRNA but not m1Ψ-mRNA. Similar immunoglobulin class-switching and somatic hypermutations were observed in response to the vaccines. Female Syrian hamsters were immunized via a prime-boost regimen with two doses of each vaccine. The titers of glycoprotein-binding antibodies were greater for U-mRNA construct than for m1Ψ-mRNA construct; however, the titers of ANDV-neutralizing antibodies were similar. Vaccinated animals were challenged with a lethal dose of ANDV, along with a naïve control group. All control animals and two animals vaccinated with a lower dose of m1Ψ-mRNA succumbed to infection whereas other vaccinated animals survived without evidence of virus replication. The data demonstrate the development of a protective vaccine against ANDV and the lack of a substantial effect of m1Ψ modification on immunogenicity and protection in rodents.


Subject(s)
Mesocricetus , Uridine , Viral Vaccines , Animals , Female , Mice , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/immunology , Antibodies, Viral/immunology , Orthohantavirus/immunology , Orthohantavirus/genetics , Antibodies, Neutralizing/immunology , Germinal Center/immunology , Pseudouridine/immunology , Cricetinae , mRNA Vaccines , Hemorrhagic Fever, American/prevention & control , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , RNA, Viral/genetics , RNA, Viral/immunology , B-Lymphocytes/immunology , Humans , Vaccine Development
16.
Front Immunol ; 15: 1433185, 2024.
Article in English | MEDLINE | ID: mdl-39081320

ABSTRACT

Tumor related angiogenesis is an attractive target in cancer therapeutic research due to its crucial role in tumor growth, invasion, and metastasis. Different agents were developed aiming to inhibit this process; however they had limited success. Cancer vaccines could be a promising tool in anti-cancer/anti-angiogenic therapy. Cancer vaccines aim to initiate an immune response against cancer cells upon presentation of tumor antigens which hopefully will result in the eradication of disease and prevention of its recurrence by inducing an efficient and long-lasting immune response. Different vaccine constructs have been developed to achieve this and they could include either protein-based or nucleic acid-based vaccines. Nucleic acid vaccines are simple and relatively easy to produce, with high efficiency and safety, thus prompting a high interest in the field. Different DNA vaccines have been developed to target crucial regulators of tumor angiogenesis. Most of them were successful in pre-clinical studies, mostly when used in combination with other therapeutics, but had limited success in the clinic. Apparently, different tumor evasion mechanisms and reduced immunogenicity still limit the potential of these vaccines and there is plenty of room for improvement. Nowadays, mRNA cancer vaccines are making remarkable progress due to improvements in the manufacturing technology and represent a powerful potential alternative. Apart from their efficiency, mRNA vaccines are simple and cheap to produce, can encompass multiple targets simultaneously, and can be quickly transferred from bench to bedside. mRNA vaccines have already accomplished amazing results in cancer clinical trials, thus ensuring a bright future in the field, although no anti-angiogenic mRNA vaccines have been described yet. This review aims to describe recent advances in anti-angiogenic DNA vaccine therapy and to provide perspectives for use of revolutionary approaches such are mRNA vaccines for anti-angiogenic treatments.


Subject(s)
Cancer Vaccines , Neoplasms , Neovascularization, Pathologic , mRNA Vaccines , Humans , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Neovascularization, Pathologic/immunology , Animals , Vaccines, DNA/immunology , Antigens, Neoplasm/immunology , Angiogenesis
17.
Hum Vaccin Immunother ; 20(1): 2382499, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39069645

ABSTRACT

Rabies, primarily transmitted to humans by dogs (accounting for 99% of cases). Once rabies occurs, its mortality rate is approximately 100%. Post-exposure prophylaxis (PEP) is critical for preventing the onset of rabies after exposure to rabid animals, and vaccination is a pivotal element of PEP. However, high costs and complex immunization protocols have led to poor adherence to rabies vaccinations. Consequently, there is an urgent need to develop new rabies vaccines that are safe, highly immunogenic, and cost-effective to improve compliance and effectively prevent rabies. In recent years, mRNA vaccines have made significant progress in the structural modification and optimization of delivery systems. Various mRNA vaccines are currently undergoing clinical trials, positioning them as viable alternatives to the traditional rabies vaccines. In this article, we discuss a novel mRNA rabies vaccine currently undergoing clinical and preclinical testing, and evaluate its potential to replace existing vaccines.


Subject(s)
Post-Exposure Prophylaxis , Rabies Vaccines , Rabies , mRNA Vaccines , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies/prevention & control , Animals , Humans , Post-Exposure Prophylaxis/methods , Rabies virus/immunology , Rabies virus/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccine Development , Dogs , Clinical Trials as Topic , RNA, Messenger/genetics , RNA, Messenger/immunology
18.
Front Biosci (Landmark Ed) ; 29(7): 246, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39082330

ABSTRACT

BACKGROUND: Pneumocystis jirovecii is the most emerging life-threating health problem that causes acute and fatal pneumonia infection. It is rare and more contagious for patients with leukemia and immune-deficiency disorders. Until now there is no treatment available for this infection therefore, it is needed to develop any treatment against this pathogen. METHODS: In this work, we used comparative proteomics, robust immune-informatics, and reverse vaccinology to create an mRNA vaccine against Pneumocystis jirovecii by targeting outer and transmembrane proteins. Using a comparative subtractive proteomic analysis of two Pneumocystis jirovecii proteomes, a distinct non-redundant Pneumocystis jirovecii (strain SE8) proteome was chosen. Seven Pneumocystis jirovecii transmembrane proteins were chosen from this proteome based on hydrophilicity, essentiality, virulence, antigenicity, pathway interaction, protein-protein network analysis, and allergenicity. OBJECTIVE: The reverse vaccinology approach was used to predict the immunogenic and antigenic epitopes of major histocompatibility complex (MHC) I, II and B-cells from the selected proteins on the basis of their antigenicity, toxicity and allergenicity. These immunogenic epitopes were linked together to construct the mRNA-based vaccine. To enhance the immunogenicity, suitable adjuvant, linkers (GPGPG, KK, and CYY), and PRDRE sequences were used. RESULTS: Through predictive modeling and confirmation via the Ramachandran plot, we assessed secondary and 3D structures. The adjuvant RpfE was incorporated to enhance the vaccine construct's immunogenicity (GRAVY index: -0.271, instability index: 39.53, antigenicity: 1.0428). The physiochemical profiling of vaccine construct was predicted it an antigenic, efficient, and potential vaccine. Notably, strong interactions were observed between the vaccine construct and TLR-3/TLR-4 (-1301.7 kcal/mol-1 and -1374.7 kcal/mol-1). CONCLUSIONS: The results predicted that mRNA-based vaccines trigger a cellular and humoral immune response, making the vaccine potential candidate against Pneumocystis jirovecii and it is more suitable for in-vitro analysis and validation to prove its effectiveness.


Subject(s)
Pneumocystis carinii , Pneumonia, Pneumocystis , Proteomics , Vaccinology , mRNA Vaccines , Proteomics/methods , Pneumocystis carinii/immunology , Pneumocystis carinii/genetics , Humans , Vaccinology/methods , mRNA Vaccines/immunology , Pneumonia, Pneumocystis/prevention & control , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/microbiology , Fungal Vaccines/immunology , Fungal Proteins/immunology , Fungal Proteins/genetics , Proteome/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccine Development/methods , Vaccines, Synthetic/immunology
19.
Vet Immunol Immunopathol ; 274: 110803, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39003921

ABSTRACT

Vaccination has proven to be an effective means of controlling pathogens in animals. Since the introduction of veterinary vaccines in the 19th century, several generations of vaccines have been introduced. These vaccines have had a positive impact on global animal health and production. Despite, the success of veterinary vaccines, there are still some pathogens for which there are no effective vaccines available, such as African swine fever. Further, animal health is under the constant threat of emerging and re-emerging pathogens, some of which are zoonotic and can pose a threat to human health. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the need for new vaccine platforms that are safe and efficacious, but also importantly, are adaptable and can be modified rapidly to match the circulating pathogens. mRNA vaccines have been shown to be an effective vaccine platform against various viral and bacterial pathogens. This review will cover some of the recent advances in the field of mRNA vaccines for veterinary species. Moreover, various mRNA vaccines and their delivery methods, as well as their reported efficacy, will be discussed. Current limitations and future prospects of this vaccine platform in veterinary medicine will also be discussed.


Subject(s)
mRNA Vaccines , Animals , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Vaccination/veterinary , RNA, Messenger/genetics
20.
Sci Transl Med ; 16(757): eadm8451, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39047117

ABSTRACT

Messenger RNA (mRNA) vaccines were pivotal in reducing severe acute respiratory syndrome 2 (SARS-CoV-2) infection burden, yet they have not demonstrated robust durability, especially in older adults. Here, we describe a molecular adjuvant comprising a lipid nanoparticle (LNP)-encapsulated mRNA encoding interleukin-12p70 (IL-12p70). The bioactive adjuvant was engineered with a multiorgan protection (MOP) sequence to restrict transcript expression to the intramuscular injection site. Admixing IL-12-MOP (CTX-1796) with the BNT162b2 SARS-CoV-2 vaccine increased spike protein-specific immune responses in mice. Specifically, the benefits of IL-12-MOP adjuvantation included amplified humoral and cellular immunity and increased immune durability for 1 year after vaccination in mice. An additional benefit included the restoration of immunity in aged mice to amounts comparable to those achieved in young adult animals, alongside amplification with a single immunization. Associated enhanced dendritic cell and germinal center responses were observed. Together, these data demonstrate that an LNP-encapsulated IL-12-MOP mRNA-encoded adjuvant can amplify immunogenicity independent of age, demonstrating translational potential to benefit vulnerable populations.


Subject(s)
Adjuvants, Immunologic , COVID-19 Vaccines , Interleukin-12 , RNA, Messenger , SARS-CoV-2 , mRNA Vaccines , Animals , Interleukin-12/metabolism , SARS-CoV-2/immunology , RNA, Messenger/metabolism , RNA, Messenger/genetics , COVID-19 Vaccines/immunology , Mice , Nanoparticles/chemistry , Female , COVID-19/prevention & control , COVID-19/immunology , BNT162 Vaccine , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Mice, Inbred C57BL , Adjuvants, Vaccine , Humans , Lipids/chemistry , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity, Cellular , Immunity, Humoral , Liposomes
SELECTION OF CITATIONS
SEARCH DETAIL