Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.573
Filter
1.
Sci Rep ; 14(1): 13865, 2024 06 15.
Article in English | MEDLINE | ID: mdl-38879684

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 had devastating consequences for human health. Despite the introduction of several vaccines, COVID-19 continues to pose a serious health risk due to emerging variants of concern. DNA vaccines gained importance during the pandemic due to their advantages such as induction of both arms of immune response, rapid development, stability, and safety profiles. Here, we report the immunogenicity and protective efficacy of a DNA vaccine encoding spike protein with D614G mutation (named pcoSpikeD614G) and define a large-scale production process. According to the in vitro studies, pcoSpikeD614G expressed abundant spike protein in HEK293T cells. After the administration of pcoSpikeD614G to BALB/c mice through intramuscular (IM) route and intradermal route using an electroporation device (ID + EP), it induced high level of anti-S1 IgG and neutralizing antibodies (P < 0.0001), strong Th1-biased immune response as shown by IgG2a polarization (P < 0.01), increase in IFN-γ levels (P < 0.01), and increment in the ratio of IFN-γ secreting CD4+ (3.78-10.19%) and CD8+ (5.24-12.51%) T cells. Challenging K18-hACE2 transgenic mice showed that pcoSpikeD614G administered through IM and ID + EP routes conferred 90-100% protection and there was no sign of pneumonia. Subsequently, pcoSpikeD614G was evaluated as a promising DNA vaccine candidate and scale-up studies were performed. Accordingly, a large-scale production process was described, including a 36 h fermentation process of E. coli DH5α cells containing pcoSpikeD614G resulting in a wet cell weight of 242 g/L and a three-step chromatography for purification of the pcoSpikeD614G DNA vaccine.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Mice, Inbred BALB C , Mutation , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccines, DNA , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Animals , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Mice , COVID-19/prevention & control , COVID-19/immunology , HEK293 Cells , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Female , Immunogenicity, Vaccine , Immunoglobulin G/blood , Immunoglobulin G/immunology
2.
Front Immunol ; 15: 1347926, 2024.
Article in English | MEDLINE | ID: mdl-38903517

ABSTRACT

Introduction: The HVTN 105 vaccine clinical trial tested four combinations of two immunogens - the DNA vaccine DNA-HIV-PT123, and the protein vaccine AIDSVAX B/E. All combinations induced substantial antibody and CD4+ T cell responses in many participants. We have now re-examined the intracellular cytokine staining flow cytometry data using the high-resolution SWIFT clustering algorithm, which is very effective for enumerating rare populations such as antigen-responsive T cells, and also determined correlations between the antibody and T cell responses. Methods: Flow cytometry samples across all the analysis batches were registered using the swiftReg registration tool, which reduces batch variation without compromising biological variation. Registered data were clustered using the SWIFT algorithm, and cluster template competition was used to identify clusters of antigen-responsive T cells and to separate these from constitutive cytokine producing cell clusters. Results: Registration strongly reduced batch variation among batches analyzed across several months. This in-depth clustering analysis identified a greater proportion of responders than the original analysis. A subset of antigen-responsive clusters producing IL-21 was identified. The cytokine patterns in each vaccine group were related to the type of vaccine - protein antigens tended to induce more cells producing IL-2 but not IFN-γ, whereas DNA vaccines tended to induce more IL-2+ IFN-γ+ CD4 T cells. Several significant correlations were identified between specific antibody responses and antigen-responsive T cell clusters. The best correlations were not necessarily observed with the strongest antibody or T cell responses. Conclusion: In the complex HVTN105 dataset, alternative analysis methods increased sensitivity of the detection of antigen-specific T cells; increased the number of identified vaccine responders; identified a small IL-21-producing T cell population; and demonstrated significant correlations between specific T cell populations and serum antibody responses. Multiple analysis strategies may be valuable for extracting the most information from large, complex studies.


Subject(s)
AIDS Vaccines , CD4-Positive T-Lymphocytes , Cytokines , Flow Cytometry , HIV Infections , Humans , AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry/methods , Cluster Analysis , HIV Infections/immunology , HIV Infections/virology , Cytokines/metabolism , Cytokines/immunology , Immunity, Humoral , HIV Antibodies/immunology , HIV Antibodies/blood , HIV-1/immunology , Vaccines, DNA/immunology , Interleukins/immunology
3.
J Exp Clin Cancer Res ; 43(1): 157, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824552

ABSTRACT

Phosphoinositide-3-kinase γ (PI3Kγ) plays a critical role in pancreatic ductal adenocarcinoma (PDA) by driving the recruitment of myeloid-derived suppressor cells (MDSC) into tumor tissues, leading to tumor growth and metastasis. MDSC also impair the efficacy of immunotherapy. In this study we verify the hypothesis that MDSC targeting, via PI3Kγ inhibition, synergizes with α-enolase (ENO1) DNA vaccination in counteracting tumor growth.Mice that received ENO1 vaccination followed by PI3Kγ inhibition had significantly smaller tumors compared to those treated with ENO1 alone or the control group, and correlated with i) increased circulating anti-ENO1 specific IgG and IFNγ secretion by T cells, ii) increased tumor infiltration of CD8+ T cells and M1-like macrophages, as well as up-modulation of T cell activation and M1-like related transcripts, iii) decreased infiltration of Treg FoxP3+ T cells, endothelial cells and pericytes, and down-modulation of the stromal compartment and T cell exhaustion gene transcription, iv) reduction of mature and neo-formed vessels, v) increased follicular helper T cell activation and vi) increased "antigen spreading", as many other tumor-associated antigens were recognized by IgG2c "cytotoxic" antibodies. PDA mouse models genetically devoid of PI3Kγ showed an increased survival and a pattern of transcripts in the tumor area similar to that of pharmacologically-inhibited PI3Kγ-proficient mice. Notably, tumor reduction was abrogated in ENO1 + PI3Kγ inhibition-treated mice in which B cells were depleted.These data highlight a novel role of PI3Kγ in B cell-dependent immunity, suggesting that PI3Kγ depletion strengthens the anti-tumor response elicited by the ENO1 DNA vaccine.


Subject(s)
Vaccines, DNA , Animals , Mice , Vaccines, DNA/pharmacology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Humans , Cell Line, Tumor , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Disease Models, Animal , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism
4.
Proc Natl Acad Sci U S A ; 121(25): e2322264121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38865265

ABSTRACT

Despite the tremendous clinical potential of nucleic acid-based vaccines, their efficacy to induce therapeutic immune response has been limited by the lack of efficient local gene delivery techniques in the human body. In this study, we develop a hydrogel-based organic electronic device (µEPO) for both transdermal delivery of nucleic acids and in vivo microarrayed cell electroporation, which is specifically oriented toward one-step transfection of DNAs in subcutaneous antigen-presenting cells (APCs) for cancer immunotherapy. The µEPO device contains an array of microneedle-shaped electrodes with pre-encapsulated dry DNAs. Upon a pressurized contact with skin tissue, the electrodes are rehydrated, electrically triggered to release DNAs, and then electroporate nearby cells, which can achieve in vivo transfection of more than 50% of the cells in the epidermal and upper dermal layer. As a proof-of-concept, the µEPO technique is employed to facilitate transdermal delivery of neoantigen genes to activate antigen-specific immune response for enhanced cancer immunotherapy based on a DNA vaccination strategy. In an ovalbumin (OVA) cancer vaccine model, we show that high-efficiency transdermal transfection of APCs with OVA-DNAs induces robust cellular and humoral immune responses, including antigen presentation and generation of IFN-γ+ cytotoxic T lymphocytes with a more than 10-fold dose sparing over existing intramuscular injection (IM) approach, and effectively inhibits tumor growth in rodent animals.


Subject(s)
Electroporation , Immunotherapy , Vaccines, DNA , Animals , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Electroporation/methods , Mice , Immunotherapy/methods , Administration, Cutaneous , Neoplasms/therapy , Neoplasms/immunology , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Ovalbumin/immunology , Ovalbumin/administration & dosage , Antigen-Presenting Cells/immunology , Female , Mice, Inbred C57BL , Humans , Vaccination/methods
5.
J Acquir Immune Defic Syndr ; 96(4): 350-360, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38916429

ABSTRACT

BACKGROUND: An effective vaccine is required to end the HIV pandemic. We evaluated the safety and immunogenicity of a DNA (DNA-HIV-PT123) vaccine with low- or high-dose bivalent (TV1.C and 1086.C glycoprotein 120) subtype C envelope protein combinations, adjuvanted with MF59 or AS01B. METHODS: HIV Vaccine Trials Network (HVTN)108 was a randomized, placebo-controlled, double-blind, phase 1/2a trial conducted in the United States and South Africa. HIV-negative adults were randomly assigned to 1 of 7 intervention arms or placebo to assess DNA prime with DNA/protein/adjuvant boosts, DNA/protein/adjuvant co-administration, and low-dose protein/adjuvant regimens. HVTN111 trial participants who received an identical regimen were also included. Outcomes included safety and immunogenicity 2 weeks and 6 months after final vaccination. RESULTS: From June 2016 to July 2018, 400 participants were enrolled (N = 334 HVTN108, N = 66 HVTN111); 370 received vaccine and 30 received placebo. There were 48 grade 3 and 3 grade 4 reactogenicity events among 39/400 (9.8%) participants, and 32 mild/moderate-related adverse events in 23/400 (5.8%) participants. All intervention groups demonstrated high IgG response rates (>89%) and high magnitudes to HIV-1 Env gp120 and gp140 proteins; response rates for AS01B-adjuvanted groups approached 100%. V1V2 IgG magnitude, Fc-mediated functions, IgG3 Env response rates, and CD4+ T-cell response magnitudes and rates were higher in the AS01B-adjuvanted groups. The AS01B-adjuvanted low-dose protein elicited greater IgG responses than the higher protein dose. CONCLUSIONS: The vaccine regimens were generally well tolerated. Co-administration of DNA with AS01B-adjuvanted bivalent Env gp120 elicited the strongest humoral responses; AS01B-adjuvanted regimens elicited stronger CD4+ T-cell responses, justifying further evaluation.ClinicalTrials.gov registration: NCT02915016, registered 26 September 2016.


Subject(s)
AIDS Vaccines , Adjuvants, Immunologic , HIV Antibodies , HIV Envelope Protein gp120 , HIV Infections , HIV-1 , Polysorbates , Squalene , Vaccines, DNA , Humans , AIDS Vaccines/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/adverse effects , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Female , Male , Adult , Squalene/administration & dosage , Polysorbates/administration & dosage , HIV Envelope Protein gp120/immunology , Adjuvants, Immunologic/administration & dosage , HIV-1/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Antibodies/blood , Double-Blind Method , Middle Aged , Young Adult , Adjuvants, Vaccine/administration & dosage , South Africa , Immunogenicity, Vaccine , Adolescent , United States
6.
J Med Virol ; 96(6): e29749, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888113

ABSTRACT

Human immunodeficiency virus (HIV) infection is still a global public health issue, and the development of an effective prophylactic vaccine inducing potent neutralizing antibodies remains a significant challenge. This study aims to explore the inflammation-related proteins associated with the neutralizing antibodies induced by the DNA/rTV vaccine. In this study, we employed the Olink chip to analyze the inflammation-related proteins in plasma in healthy individuals receiving HIV candidate vaccine (DNA priming and recombinant vaccinia virus rTV boosting) and compared the differences between neutralizing antibody-positive (nab + ) and -negative(nab-) groups. We identified 25 differentially expressed factors and conducted enrichment and correlation analysis on them. Our results revealed that significant expression differences in artemin (ARTN) and C-C motif chemokine ligand 23 (CCL23) between nab+ and -nab- groups. Notably, the expression of CCL23 was negatively corelated to the ID50 of neutralizing antibodies and the intensity of the CD4+ T cell responses. This study enriches our understanding of the immune picture induced by the DNA/rTV vaccine, and provides insights for future HIV vaccine development.


Subject(s)
AIDS Vaccines , Antibodies, Neutralizing , HIV Antibodies , HIV Infections , HIV-1 , Proteomics , Vaccinia virus , Humans , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Vaccinia virus/immunology , Vaccinia virus/genetics , HIV Antibodies/blood , HIV Antibodies/immunology , HIV-1/immunology , HIV-1/genetics , Adult , AIDS Vaccines/immunology , Male , HIV Infections/immunology , Vaccines, DNA/immunology , Female , Healthy Volunteers , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Plasma/immunology , Young Adult
7.
Antiviral Res ; 227: 105914, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38759930

ABSTRACT

Due to the severity of CMV infection in immunocompromised individuals the development of a vaccine has been declared a priority. However, despite the efforts made there is no yet a vaccine available for clinical use. We designed an approach to identify new CMV antigens able to inducing a broad immune response that could be used in future vaccine formulations. We have used serum samples from 28 kidney transplant recipients, with a previously acquired CMV-specific immune response to identify viral proteins that were recognized by the antibodies present in the patient serum samples by Western blot. A band of approximately 45 kDa, identified as UL44, was detected by most serum samples. UL44 immunogenicity was tested in BALB/c mice that received three doses of the UL44-pcDNA DNA vaccine. UL44 elicited both, a strong antibody response and CMV-specific cellular response. Using bioinformatic analysis we demonstrated that UL44 is a highly conserved protein and contains epitopes that are able to activate CD8 lymphocytes of the most common HLA alleles in the world population. We constructed a UL44 ORF deletion mutant virus that produced no viral progeny, suggesting that UL44 is an essential viral protein. In addition, other authors have demonstrated that UL44 is one of the most abundant viral proteins after infection and have suggested an essential role of UL44 in viral replication. Altogether, our data suggests that UL44 is a potent antigen, and favored by its abundance, it may be a good candidate to include in a vaccine formulation.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Mice, Inbred BALB C , Viral Proteins , Animals , Mice , Humans , Cytomegalovirus/immunology , Cytomegalovirus/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Viral Proteins/immunology , Viral Proteins/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Female , Cytomegalovirus Vaccines/immunology , Cytomegalovirus Vaccines/administration & dosage , T-Lymphocytes/immunology , Antigens, Viral/immunology , Kidney Transplantation , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular
8.
Fish Shellfish Immunol ; 150: 109663, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38821228

ABSTRACT

Persistent nocardiosis has prompted exploration of the effectiveness of heterologous approaches to prevent severe infections. We have previously reported the efficacy of a nucleic acid vaccine in protecting groupers from highly virulent Nocardia seriolae infections. Ongoing research has involved the supplementation of recombinant cholesterol oxidase (rCho) proteins through immunization with a DNA vaccine to enhance the protective capacity of orange-spotted groupers. Recombinant rCho protein exhibited a maturity and biological structure comparable to that expressed in N. seriolae, as confirmed by Western blot immunodetection assays. The immune responses observed in vaccinated groupers were significantly higher than those observed in single-type homologous vaccinations, DNA or recombinant proteins alone (pcD:Cho and rCho/rCho), especially cell-mediated immune and mucosal immune responses. Moreover, the reduction in N. seriolae occurrence in internal organs, such as the head, kidney, and spleen, was consistent with the vaccine's efficacy, which increased from approximately 71.4 % to an undetermined higher percentage through heterologous vaccination strategies of 85.7 %. This study underscores the potential of Cho as a novel vaccine candidate and a heterologous approach for combating chronic infections such as nocardiosis.


Subject(s)
Bacterial Vaccines , Fish Diseases , Nocardia Infections , Nocardia , Animals , Nocardia Infections/veterinary , Nocardia Infections/prevention & control , Nocardia Infections/immunology , Nocardia/immunology , Fish Diseases/prevention & control , Fish Diseases/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Bass/immunology , Cholesterol Oxidase/immunology , Cholesterol Oxidase/genetics , Recombinant Proteins/immunology , Recombinant Proteins/administration & dosage
9.
J Immunother Cancer ; 12(5)2024 May 20.
Article in English | MEDLINE | ID: mdl-38772685

ABSTRACT

RATIONALE: Androgen deprivation therapy (ADT) is the primary treatment for recurrent and metastatic prostate cancer. In addition to direct antitumor effects, ADT has immunomodulatory effects such as promoting T-cell infiltration and enhancing antigen processing/presentation. Previous studies in our laboratory have demonstrated that ADT also leads to increased expression of the androgen receptor (AR) and increased recognition of prostate tumor cells by AR-specific CD8+T cells. We have also demonstrated that ADT combined with a DNA vaccine encoding the AR significantly slowed tumor growth and improved the survival of prostate tumor-bearing mice. The current study aimed to investigate the impact of the timing and sequencing of ADT with vaccination on the tumor immune microenvironment in murine prostate cancer models to further increase the antitumor efficacy of vaccines. METHODS: Male FVB mice implanted with Myc-CaP tumor cells, or male C57BL/6 mice implanted with TRAMP-C1 prostate tumor cells, were treated with a DNA vaccine encoding AR (pTVG-AR) and ADT. The sequence of administration was evaluated for its effect on tumor growth, and tumor-infiltrating immune populations were characterized. RESULTS: Vaccination prior to ADT (pTVG-AR → ADT) significantly enhanced antitumor responses and survival. This was associated with increased tumor infiltration by CD4+ and CD8+ T cells, including AR-specific CD8+T cells. Depletion of CD8+T cells prior to ADT significantly worsened overall survival. Following ADT treatment, however, Gr1+ myeloid-derived suppressor cells (MDSCs) increased, and this was associated with fewer infiltrating T cells and reduced tumor growth. Inhibiting Gr1+MDSCs recruitment, either by using a CXCR2 antagonist or by cycling androgen deprivation with testosterone replacement, improved antitumor responses and overall survival. CONCLUSION: Vaccination prior to ADT significantly improved antitumor responses, mediated in part by increased infiltration of CD8+T cells following ADT. Targeting MDSC recruitment following ADT further enhanced antitumor responses. These findings suggest logical directions for future clinical trials to improve the efficacy of prostate cancer vaccines.


Subject(s)
Cancer Vaccines , Prostatic Neoplasms , Receptors, Androgen , Male , Animals , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Cancer Vaccines/therapeutic use , Cancer Vaccines/pharmacology , Cancer Vaccines/immunology , Vaccines, DNA/therapeutic use , Vaccines, DNA/pharmacology , Androgen Antagonists/therapeutic use , Androgen Antagonists/pharmacology , Cell Line, Tumor , Mice, Inbred C57BL , Vaccination , Humans , Tumor Microenvironment , Disease Models, Animal , CD8-Positive T-Lymphocytes/immunology
10.
Vaccine ; 42(17): 3733-3743, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38705805

ABSTRACT

Hand, foot, and mouth disease (HFMD) poses a significant public health threat primarily caused by four major enteroviruses: enterovirus 71 (EV71), coxsackieviruses A16, A10, and A6. Broadly protective immune responses are essential for complete protection against these major enteroviruses. In this study, we designed a new tetravalent immunogen for HFMD, validated it in silico, in vivo evaluated the immunogenicity of the DNA-based tetravalent vaccine in mice, and identified immunogenic B-cell and T-cell epitopes. A new tetravalent immunogen, VP1me, was designed based on the chimeric protein and epitope-based vaccine principles. It contains a complete EV71 VP1 protein and six reported neutralizing B-cell epitopes derived from the four major enteroviruses causing HFMD. In silico validation using multiple immunoinformatic tools indicated good attributes of the VP1me immunogen suitable for vaccine development. The VP1me-based DNA vaccine efficiently induced both humoral and cellular immune responses in BALB/cAJcl mice. A combination of in silico prediction and immunoassays enabled the identification of immunogenic linear B-cell and CD8 T-cell epitopes within the VP1me immunogen. Immunodominant linear B-cell epitopes were identified in six regions of VP1me, with one epitope located at the N-terminus of the VP1 protein (aa 9-23) regarded as a novel epitope. Interestingly, some B-cell epitopes could also induce the CD8 T-cell response, suggesting their dual functions in immune stimulation. These results lay the groundwork for further development of VP1me as a new vaccine candidate.


Subject(s)
Antibodies, Viral , Epitopes, B-Lymphocyte , Hand, Foot and Mouth Disease , Immunodominant Epitopes , Mice, Inbred BALB C , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Epitopes, B-Lymphocyte/immunology , Hand, Foot and Mouth Disease/prevention & control , Hand, Foot and Mouth Disease/immunology , Mice , Viral Vaccines/immunology , Immunodominant Epitopes/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Female , Epitopes, T-Lymphocyte/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Enterovirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Enterovirus A, Human/immunology , Enterovirus A, Human/genetics , Immunogenicity, Vaccine , Immunity, Cellular , Immunity, Humoral
11.
Nat Commun ; 15(1): 4301, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773089

ABSTRACT

The vaccine elicitation of HIV tier-2-neutralization antibodies has been a challenge. Here, we report the isolation and characterization of a CD4-binding site (CD4bs) specific monoclonal antibody, HmAb64, from a human volunteer immunized with a polyvalent DNA prime-protein boost HIV vaccine. HmAb64 is derived from heavy chain variable germline gene IGHV1-18 and light chain germline gene IGKV1-39. It has a third heavy chain complementarity-determining region (CDR H3) of 15 amino acids. On a cross-clade panel of 208 HIV-1 pseudo-virus strains, HmAb64 neutralized 20 (10%), including tier-2 strains from clades B, BC, C, and G. The cryo-EM structure of the antigen-binding fragment of HmAb64 in complex with a CNE40 SOSIP trimer revealed details of its recognition; HmAb64 uses both heavy and light CDR3s to recognize the CD4-binding loop, a critical component of the CD4bs. This study demonstrates that a gp120-based vaccine can elicit antibodies capable of tier 2-HIV neutralization.


Subject(s)
AIDS Vaccines , Antibodies, Neutralizing , CD4 Antigens , HIV Antibodies , HIV-1 , Humans , AIDS Vaccines/immunology , HIV-1/immunology , HIV Antibodies/immunology , Antibodies, Neutralizing/immunology , CD4 Antigens/immunology , CD4 Antigens/metabolism , Vaccines, DNA/immunology , Antibodies, Monoclonal/immunology , HIV Infections/prevention & control , HIV Infections/immunology , HIV Infections/virology , Cryoelectron Microscopy , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/chemistry , Binding Sites , Complementarity Determining Regions/immunology , Complementarity Determining Regions/chemistry
12.
BMC Infect Dis ; 24(1): 476, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714948

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne viral disease caused by the SFTS virus (Dabie bandavirus), which has become a substantial risk to public health. No specific treatment is available now, that calls for an effective vaccine. Given this, we aimed to develop a multi-epitope DNA vaccine through the help of bioinformatics. The final DNA vaccine was inserted into a special plasmid vector pVAX1, consisting of CD8+ T cell epitopes, CD4+ T cell epitopes and B cell epitopes (six epitopes each) screened from four genome-encoded proteins--nuclear protein (NP), glycoprotein (GP), RNA-dependent RNA polymerase (RdRp), as well as nonstructural protein (NSs). To ascertain if the predicted structure would be stable and successful in preventing infection, an immunological simulation was run on it. In conclusion, we designed a multi-epitope DNA vaccine that is expected to be effective against Dabie bandavirus, but in vivo trials are needed to verify this claim.


Subject(s)
Epitopes, T-Lymphocyte , Phlebovirus , Severe Fever with Thrombocytopenia Syndrome , Vaccines, DNA , Viral Vaccines , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Phlebovirus/immunology , Phlebovirus/genetics , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Severe Fever with Thrombocytopenia Syndrome/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Humans , Computer-Aided Design , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Animals , Computational Biology
14.
Lancet HIV ; 11(5): e285-e299, 2024 May.
Article in English | MEDLINE | ID: mdl-38692824

ABSTRACT

BACKGROUND: An effective HIV vaccine will most likely need to have potent immunogenicity and broad cross-subtype coverage. The aim of the HIV Vaccine Trials Network (HVTN) 124 was to evaluate safety and immunogenicity of a unique polyvalent DNA-protein HIV vaccine with matching envelope (Env) immunogens. METHODS: HVTN 124 was a randomised, phase 1, placebo-controlled, double-blind study, including participants who were HIV seronegative and aged 18-50 years at low risk for infection. The DNA vaccine comprised five plasmids: four copies expressing Env gp120 (clades A, B, C, and AE) and one gag p55 (clade C). The protein vaccine included four DNA vaccine-matched GLA-SE-adjuvanted recombinant gp120 proteins. Participants were enrolled across six clinical sites in the USA and were randomly assigned to placebo or one of two vaccine groups (ie, prime-boost or coadministration) in a 5:1 ratio in part A and a 7:1 ratio in part B. Vaccines were delivered via intramuscular needle injection. The primary outcomes were safety and tolerability, assessed via frequency, severity, and attributability of local and systemic reactogenicity and adverse events, laboratory safety measures, and early discontinuations. Part A evaluated safety. Part B evaluated safety and immunogenicity of two regimens: DNA prime (administered at months 0, 1, and 3) with protein boost (months 6 and 8), and DNA-protein coadministration (months 0, 1, 3, 6, and 8). All randomly assigned participants who received at least one dose were included in the safety analysis. The study is registered with ClinicalTrials.gov (NCT03409276) and is closed to new participants. FINDINGS: Between April 19, 2018 and Feb 13, 2019, 60 participants (12 in part A [five men and seven women] and 48 in part B [21 men and 27 women]) were enrolled. All 60 participants received at least one dose, and 14 did not complete follow-up (six of 21 in the prime-boost group and eight of 21 in the coadminstration group). 11 clinical adverse events deemed by investigators as study-related occurred in seven of 48 participants in part B (eight of 21 in the prime-boost group and three of 21 in the coadministration group). Local reactogenicity in the vaccine groups was common, but the frequency and severity of reactogenicity signs or symptoms did not differ between the prime-boost and coadministration groups (eg, 20 [95%] of 21 in the prime-boost group vs 21 [100%] of 21 in the coadministration group had either local pain or tenderness of any severity [p=1·00], and seven [33%] vs nine [43%] had either erythema or induration [p=0·97]), nor did laboratory safety measures. There were no delayed-type hypersensitivity reactions or vasculitis or any severe clinical adverse events related to vaccination. The most frequently reported systemic reactogenicity symptoms in the active vaccine groups were malaise or fatigue (five [50%] of ten in part A and 17 [81%] of 21 in the prime-boost group vs 15 [71%] of 21 in the coadministration group in part B), headache (five [50%] and 18 [86%] vs 12 [57%]), and myalgia (four [40%] and 13 [62%] vs ten [48%]), mostly of mild or moderate severity. INTERPRETATION: Both vaccine regimens were safe, warranting evaluation in larger trials. FUNDING: US National Institutes of Health and US National Institute of Allergy and Infectious Diseases.


Subject(s)
AIDS Vaccines , HIV Antibodies , HIV Infections , HIV-1 , Vaccines, DNA , Humans , AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , AIDS Vaccines/adverse effects , Adult , Male , Female , Double-Blind Method , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, DNA/adverse effects , HIV Infections/prevention & control , HIV Infections/immunology , Middle Aged , Young Adult , HIV Antibodies/blood , Adolescent , HIV-1/immunology , United States , Immunization, Secondary , Immunogenicity, Vaccine , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/genetics , Antibodies, Neutralizing/blood
15.
Mol Biochem Parasitol ; 259: 111630, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38795969

ABSTRACT

Toxoplasma gondii is an intracellular protozoan parasite that infects all nucleated cells except the red blood cells. Currently, nucleic acid vaccines are being widely investigated in Toxoplasma gondii control, and several nucleic acid vaccine candidate antigens have shown good protection in various studies. The aim of this study was to construct a nucleic acid vaccine with Toxoplasma gondii SRS29C as the target gene. We explored the nucleic acid vaccine with Toxoplasma surface protein SRS29C and the combined gene of SRS29C and SAG1 and evaluated its immunoprotective effect against Toxoplasma gondii. To amplify the gene fragment and clone it to the expression vector, the recombinant plasmid pEGFP-SRS29C was constructed by PCR. Eukaryotic cells were transfected with the plasmid, and the expression of the target protein was assessed using the Western blot method. The level of serum IgG was determined via ELISA, and the splenic lymphocyte proliferation ability was detected using the CCK-8 method. The percentages of CD4+ and CD8+ T cells were measured by flow cytometry. Mice were immunised three times with single-gene nucleic acid vaccine and combination vaccine. Splenic lymphocytokine expression was determined using ELISA kits. The mice's survival time was monitored and recorded during an in vivo insect assault experiment, and the vaccine's protective power was assessed. The outcomes showed that PCR-amplification of an SRS29C gene fragment was successful. The 4,733-bp vector fragment and the 1,119-bp target segment were both recognised by double digestion. Additionally, after transfection of the recombinant plasmid pEGFP-SRS29C, Western blot examination of the extracted protein revealed the presence of a target protein strip at 66 kDa. The test results demonstrated that the IgG content in the serum of the pEGFP-SRS29C group and the co-immunization group was significantly higher than that of the PBS group and the empty vector group. The IgG potency induced by the co-immunization group was higher than that of the pEGFP-SRS29C group and the pEGFP-SAG1 group, the number of splenic lymphocyte proliferation number was higher than that of the PBS group and the empty vector group. The CD4+/CD8+ T ratio was higher than that of the PBS group and the empty vector group. The expression of IFN-γ and TNF-α in the splenocytes of the pEGFP-SRS29C group and the combined immunisation group was significantly higher following antigen stimulation. In the worm attack experiments, mice in the PBS and empty vector groups perished within 9 days of the worm attack, whereas mice in the pEGFP-SRS29C group survived for 18 days, mice in the pEGFP-SAG1 group survived for 21 days, and mice in the co-immunization group survived for 24 days. This demonstrates that the constructed Toxoplasma gondii nucleic acid vaccine pEGFP-SRS29C and the combined gene vaccine can induce mice to develop certain humoral and cellular immune responses, and enhance their ability to resist Toxoplasma gondii infection.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Immunoglobulin G , Protozoan Proteins , Protozoan Vaccines , Toxoplasma , Vaccines, DNA , Animals , Toxoplasma/immunology , Toxoplasma/genetics , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Vaccines, DNA/administration & dosage , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Protozoan Vaccines/immunology , Protozoan Vaccines/genetics , Mice , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Immunoglobulin G/blood , Immunoglobulin G/immunology , Female , Toxoplasmosis, Animal/prevention & control , Toxoplasmosis, Animal/immunology , Mice, Inbred BALB C , CD8-Positive T-Lymphocytes/immunology , Spleen/immunology , Spleen/parasitology , Cell Proliferation , Plasmids/genetics , Plasmids/immunology , Cytokines/metabolism
16.
Adv Drug Deliv Rev ; 210: 115340, 2024 07.
Article in English | MEDLINE | ID: mdl-38810703

ABSTRACT

Nucleic acid technology has revolutionized vaccine development, enabling rapid design and production of RNA and DNA vaccines for prevention and treatment of diseases. The successful deployment of mRNA and plasmid DNA vaccines against COVID-19 has further validated the technology. At present, mRNA platform is prevailing due to its higher efficacy, while DNA platform is undergoing rapid evolution because it possesses unique advantages that can potentially overcome the problems associated with the mRNA platform. To help understand the recent performances of the two vaccine platforms and recognize their clinical potentials in the future, this review compares the advantages and drawbacks of mRNA and DNA vaccines that are currently known in the literature, in terms of development timeline, financial cost, ease of distribution, efficacy, safety, and regulatory approval of products. Additionally, the review discusses the ongoing clinical trials, strategies for improvement, and alternative designs of RNA and DNA platforms for vaccination.


Subject(s)
COVID-19 Vaccines , COVID-19 , Vaccines, DNA , mRNA Vaccines , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Humans , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Animals
17.
Hum Vaccin Immunother ; 20(1): 2352908, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38780076

ABSTRACT

Cervical cancer, among the deadliest cancers affecting women globally, primarily arises from persistent infection with high-risk human papillomavirus (HPV). To effectively combat persistent infection and prevent the progression of precancerous lesions into malignancy, a therapeutic HPV vaccine is under development. This study utilized an immunoinformatics approach to predict epitopes of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) using the E6 and E7 oncoproteins of the HPV16 strain as target antigens. Subsequently, through meticulous selection of T-cell epitopes and other necessary elements, a multi-epitope vaccine was constructed, exhibiting good immunogenic, physicochemical, and structural characteristics. Furthermore, in silico simulations showed that the vaccine not only interacted well with toll-like receptors (TLR2/TLR3/TLR4), but also induced a strong innate and adaptive immune response characterized by elevated Th1-type cytokines, such as interferon-gamma (IFN-γ) and interleukin-2 (IL2). Additionally, our study investigated the effects of different immunization intervals on immune responses, aiming to optimize a time-efficient immunization program. In animal model experiments, the vaccine exhibited robust immunogenic, therapeutic, and prophylactic effects. Administered thrice, it consistently induced the expansion of specific CD4 and CD8 T cells, resulting in substantial cytokines release and increased proliferation of memory T cell subsets in splenic cells. Overall, our findings support the potential of this multi-epitope vaccine in combating HPV16 infection and signify its candidacy for future HPV vaccine development.


Through the stringent selection of T-cell epitopes and other necessary elements, a novel multi-epitope vaccine targeting HPV 16 E6 and E7 oncoproteins was constructed using an immunoinformatics approach.The vaccine designed can induce both cellular and humoral immune responses, encompassing all the required immunogenic, physicochemical, and structural characteristics for an ideal vaccine design. Moreover, it offers decent worldwide coverage.In animal studies, the vaccine demonstrated strong immune responses, including expansion of CD4 and CD8 T cells, cytokine release, and enhanced memory T cell proliferation, resulting in long-term anti-tumor effects, inhibition of tumor growth, and prolonged survival in tumor-bearing mice.The immunological evaluation of the designed vaccine suggests its potential as a novel vaccine candidate against HPV 16.


Subject(s)
Epitopes, T-Lymphocyte , Human papillomavirus 16 , Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Vaccines, DNA , Female , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Human papillomavirus 16/immunology , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Epitopes, T-Lymphocyte/immunology , Animals , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Papillomavirus E7 Proteins/immunology , Mice , Humans , T-Lymphocytes, Cytotoxic/immunology , Repressor Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Mice, Inbred C57BL , Interferon-gamma/metabolism , Interferon-gamma/immunology
18.
Methods Mol Biol ; 2786: 303-319, 2024.
Article in English | MEDLINE | ID: mdl-38814401

ABSTRACT

A plasmid production process has been established to manufacture plasmid DNA at a large scale in High-Quality grade. This is used as a starting material to produce mRNA vaccines for clinical trials. Recently, the World Health Organization (WHO) has released regulatory guidelines related to the quality, safety, and efficacy for DNA- as well as for mRNA-based vaccines. Following an extraordinary year of scientific, regulatory, and manufacturing developments, the scientific community today stands considerably better equipped to deal with urgent production requirements in large scale for nucleic acid-based vaccinations and therapies. Going forward, work needs to be done in better coordinating the supply and logistics of essential raw materials for biological manufacturing, especially under emergency conditions.


Subject(s)
Plasmids , Vaccines, DNA , Plasmids/genetics , Humans , Vaccines, DNA/genetics , Vaccines, DNA/immunology , mRNA Vaccines
19.
Virol Sin ; 39(3): 490-500, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38768713

ABSTRACT

As of December 2022, 2603 laboratory-identified Middle East respiratory syndrome coronavirus (MERS-CoV) infections and 935 associated deaths, with a mortality rate of 36%, had been reported to the World Health Organization (WHO). However, there are still no vaccines for MERS-CoV, which makes the prevention and control of MERS-CoV difficult. In this study, we generated two DNA vaccine candidates by integrating MERS-CoV Spike (S) gene into a replicating Vaccinia Tian Tan (VTT) vector. Compared to homologous immunization with either vaccine, mice immunized with DNA vaccine prime and VTT vaccine boost exhibited much stronger and durable humoral and cellular immune responses. The immunized mice produced robust binding antibodies and broad neutralizing antibodies against the EMC2012, England1 and KNIH strains of MERS-CoV. Prime-Boost immunization also induced strong MERS-S specific T cells responses, with high memory and poly-functional (CD107a-IFN-γ-TNF-α) effector CD8+ T cells. In conclusion, the research demonstrated that DNA-Prime/VTT-Boost strategy could elicit robust and balanced humoral and cellular immune responses against MERS-CoV-S. This study not only provides a promising set of MERS-CoV vaccine candidates, but also proposes a heterologous sequential immunization strategy worthy of further development.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Coronavirus Infections , Immunity, Cellular , Immunity, Humoral , Mice, Inbred BALB C , Middle East Respiratory Syndrome Coronavirus , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Middle East Respiratory Syndrome Coronavirus/immunology , Middle East Respiratory Syndrome Coronavirus/genetics , Antibodies, Viral/blood , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Female , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Immunization, Secondary , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics
20.
Hum Vaccin Immunother ; 20(1): 2346390, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38691025

ABSTRACT

Middle East respiratory coronavirus (MERS-CoV) is a newly emergent, highly pathogenic coronavirus that is associated with 34% mortality rate. MERS-CoV remains listed as priority pathogen by the WHO. Since its discovery in 2012 and despite the efforts to develop coronaviruses vaccines to fight against SARS-CoV-2, there are currently no MERS-CoV vaccine that has been approved. Therefore, there is high demand to continue on the development of prophylactic vaccines against MERS-CoV. Current advancements in vaccine developments can be adapted for the development of improved MERS-CoV vaccines candidates. Nucleic acid-based vaccines, including pDNA and mRNA, are relatively new class of vaccine platforms. In this work, we developed pDNA and mRNA vaccine candidates expressing S.FL gene of MERS-CoV. Further, we synthesized a silane functionalized hierarchical aluminosilicate to encapsulate each vaccine candidates. We tested the nucleic acid vaccine candidates in mice and evaluated humoral antibodies response. Interestingly, we determined that the non-encapsulated, codon optimized S.FL pDNA vaccine candidate elicited the highest level of antibody responses against S.FL and S1 of MERS-CoV. Encapsulation of mRNA with nanoporous aluminosilicate increased the humoral antibody responses, whereas encapsulation of pDNA did not. These findings suggests that MERS-CoV S.FL pDNA vaccine candidate induced the highest level of humoral responses. This study will enhance further optimization of nanosilica as potential carrier for mRNA vaccines. In conclusion, this study suggests MERS-CoV pDNA vaccine candidate as a suitable vaccine platform for further pivotal preclinical testings.


Subject(s)
Antibodies, Viral , Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , Nanoparticles , Silicon Dioxide , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Vaccines, DNA/administration & dosage , Middle East Respiratory Syndrome Coronavirus/immunology , Middle East Respiratory Syndrome Coronavirus/genetics , Mice , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Antibodies, Viral/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Silicon Dioxide/chemistry , Mice, Inbred BALB C , Female , Humans , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Vaccine Development
SELECTION OF CITATIONS
SEARCH DETAIL
...