1
|
Bose D, Rogers KA, Shirreff LM, Chebloune Y, Villinger FJ. Enhanced HIV immune responses elicited by an apoptotic single-cycle SHIV lentivector DNA vaccine. Front Cell Infect Microbiol 2025; 15:1481427. [PMID: 40292218 PMCID: PMC12023480 DOI: 10.3389/fcimb.2025.1481427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
Background HIV remains a major public health issue in spite of antiretroviral therapy (ART). An innovative vaccine that can induce long-lasting and effective immunity is required to curb the persistently high numbers of new infections worldwide. Methods A novel DNA vaccine was generated using a Simian-Human Immunodeficiency Virus (SHIV) backbone with a Zambian T/F clade C envelope and under the control of the caprine arthritis encephalitis virus long terminal repeats (LTRs) for constitutive expression. Due to the deleted integrase, this DNA vaccine "CSH-DIN-T/F Z331" performs only a single replication cycle. To increase immunogenicity, the co-expression of apoptotic genes (BAX, BAK, or caspase 8) incorporated at the end of Pol was tested to promote the release of apoptotic bodies taken up by dendritic cells leading to cross-presentation of antigen. The three vaccines (CSH-DIN-T/F Z331-BAX, CSH-DIN-T/F Z331-BAK, and CSH-DIN-T/F Z331-Cas8) were tested in vitro for expression and in vivo in BALB/cJ mice for immunogenicity. Results Transduced HEK293 cells co-cultured with CEMx174 confirmed the single replication cycle of the DNA vaccine and the induction of apoptosis by CSH-DIN-T/F Z331-Cas8 based on Annexin V expression. BALB/cJ mice were immunized with a combined intramuscular + intradermal/electroporation approach. Intracellular cytokine staining (ICS) from splenocytes collected 12 weeks post-prime/6 weeks post-boost demonstrated a clear superiority of caspase 8 expressing construct over the others, with higher proportions of IFN-γ-, IL-2-, and IL-21-producing CD8 T cells specific to Env, Gag, and Nef. The kinetics of immune response after various immunization schedules were also investigated. Conclusion This novel single-cycle DNA vaccine with apoptotic genes demonstrated an enhanced immunogenicity primarily for antigen-specific CD8+ T-cell responses.
Collapse
Affiliation(s)
- Deepanwita Bose
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, United States
| | - Kenneth A. Rogers
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, United States
| | - Lisa M. Shirreff
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, United States
| | - Yahia Chebloune
- Laboratoire Pathogénèse et Vaccination Lentivirales (PAVAL) Lab., Institut National de Recherche d’Agriculture et Environnement, Université Grenoble Alpes, Saint Martin d’Hères, France
| | - Francois J. Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, United States
| |
Collapse
|
2
|
Vaccination with Combination DNA and Virus-Like Particles Enhances Humoral and Cellular Immune Responses upon Boost with Recombinant Modified Vaccinia Virus Ankara Expressing Human Immunodeficiency Virus Envelope Proteins. Vaccines (Basel) 2017; 5:vaccines5040052. [PMID: 29257056 PMCID: PMC5748618 DOI: 10.3390/vaccines5040052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/06/2017] [Accepted: 12/12/2017] [Indexed: 01/13/2023] Open
Abstract
Heterologous prime boost with DNA and recombinant modified vaccinia virus Ankara (rMVA) vaccines is considered as a promising vaccination approach against human immunodeficiency virus (HIV-1). To further enhance the efficacy of DNA-rMVA vaccination, we investigated humoral and cellular immune responses in mice after three sequential immunizations with DNA, a combination of DNA and virus-like particles (VLP), and rMVA expressing HIV-1 89.6 gp120 envelope proteins (Env). DNA prime and boost with a combination of VLP and DNA vaccines followed by an rMVA boost induced over a 100-fold increase in Env-specific IgG antibody titers compared to three sequential immunizations with DNA and rMVA. Cellular immune responses were induced by VLP-DNA and rMVA vaccinations at high levels in CD8 T cells, CD4 T cells, and peripheral blood mononuclear cells secreting interferon (IFN)-γ, and spleen cells producing interleukin (IL)-2, 4, 5 cytokines. This study suggests that a DNA and VLP combination vaccine with MVA is a promising strategy in enhancing the efficacy of DNA-rMVA vaccination against HIV-1.
Collapse
|
3
|
Chea LS, Amara RR. Immunogenicity and efficacy of DNA/MVA HIV vaccines in rhesus macaque models. Expert Rev Vaccines 2017; 16:973-985. [PMID: 28838267 DOI: 10.1080/14760584.2017.1371594] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Despite 30 years of research on HIV, a vaccine to prevent infection and limit disease progression remains elusive. The RV144 trial showed moderate, but significant protection in humans and highlighted the contribution of antibody responses directed against HIV envelope as an important immune correlate for protection. Efforts to further build upon the progress include the use of a heterologous prime-boost regimen using DNA as the priming agent and the attenuated vaccinia virus, Modified Vaccinia Ankara (MVA), as a boosting vector for generating protective HIV-specific immunity. Areas covered: In this review, we summarize the immunogenicity of DNA/MVA vaccines in non-human primate models and describe the efficacy seen in SIV infection models. We discuss immunological correlates of protection determined by these studies and potential approaches for improving the protective immunity. Additionally, we describe the current progress of DNA/MVA vaccines in human trials. Expert commentary: Efforts over the past decade have provided the opportunity to better understand the dynamics of vaccine-induced immune responses and immune correlates of protection against HIV. Based on what we have learned, we outline multiple areas where the field will likely focus on in the next five years.
Collapse
Affiliation(s)
- Lynette Siv Chea
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| | - Rama Rao Amara
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| |
Collapse
|
4
|
Chamcha V, Jones A, Quigley BR, Scott JR, Amara RR. Oral Immunization with a Recombinant Lactococcus lactis-Expressing HIV-1 Antigen on Group A Streptococcus Pilus Induces Strong Mucosal Immunity in the Gut. THE JOURNAL OF IMMUNOLOGY 2015; 195:5025-34. [PMID: 26482408 DOI: 10.4049/jimmunol.1501243] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/16/2015] [Indexed: 01/25/2023]
Abstract
The induction of a potent humoral and cellular immune response in mucosal tissue is important for the development of an effective HIV vaccine. Most of the current HIV vaccines under development use the i.m. route for immunization, which is relatively poor in generating potent and long-lived mucosal immune responses. In this article, we explore the ability of an oral vaccination with a probiotic organism, Lactococcus lactis, to elicit HIV-specific immune responses in the mucosal and systemic compartments of BALB/c mice. We expressed the HIV-1 Gag-p24 on the tip of the T3 pilus of Streptococcus pyogenes as a fusion to the Cpa protein (LL-Gag). After four monthly LL-Gag oral immunizations, we observed strong Gag-specific IgG and IgA responses in serum, feces, and vaginal secretions. However, the Gag-specific CD8 T cell responses in the blood were at or below our detection limit. After an i.m. modified vaccinia Ankara/Gag boost, we observed robust Gag-specific CD8 T cell responses both in systemic and in mucosal tissues, including intraepithelial and lamina propria lymphocytes of the small intestine, Peyer's patches, and mesenteric lymph nodes. Consistent with strong immunogenicity, the LL-Gag induced activation of CD11c(+) CD11b(+) dendritic cells in the Peyer's patches after oral immunization. Our results demonstrate that oral immunization with L. lactis expressing an Ag on the tip of the group A Streptococcus pilus serves as an excellent vaccine platform to induce strong mucosal humoral and cellular immunity against HIV.
Collapse
Affiliation(s)
- Venkateswarlu Chamcha
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Andrew Jones
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Bernard R Quigley
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30329
| | - June R Scott
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30329
| | - Rama Rao Amara
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and Department of Microbiology and Immunology, Emory University, Atlanta, GA 30329
| |
Collapse
|
5
|
Xu J, Zhang Y, Tao J. Efficacy of a DNA vaccine carrying Eimeria maxima Gam56 antigen gene against coccidiosis in chickens. THE KOREAN JOURNAL OF PARASITOLOGY 2013; 51:147-54. [PMID: 23710081 PMCID: PMC3662057 DOI: 10.3347/kjp.2013.51.2.147] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Revised: 09/18/2012] [Accepted: 10/03/2012] [Indexed: 11/23/2022]
Abstract
To control coccidiosis without using prophylactic medications, a DNA vaccine targeting the gametophyte antigen Gam56 from Eimeria maxima in chickens was constructed, and the immunogenicity and protective effects were evaluated. The ORF of Gam56 gene was cloned into an eukaryotic expression vector pcDNA3.1(zeo)+. Expression of Gam56 protein in COS-7 cells transfected with recombinant plasmid pcDNA-Gam56 was confirmed by indirect immunofluorescence assay. The DNA vaccine was injected intramuscularly to yellow feathered broilers of 1-week old at 3 dosages (25, 50, and 100 µg/chick). Injection was repeated once 1 week later. One week after the second injection, birds were challenged orally with 5×10(4) sporulated oocysts of E. maxima, then weighed and killed at day 8 post challenge. Blood samples were collected and examined for specific peripheral blood lymphocyte proliferation activity and serum antibody levels. Compared with control groups, the administration of pcDNA-Gam56 vaccine markedly increased the lymphocyte proliferation activity (P<0.05) at day 7 and 14 after the first immunization. The level of lymphocyte proliferation started to decrease on day 21 after the first immunization. A similar trend was seen in specific antibody levels. Among the 3 pcDNA-Gam56 immunized groups, the median dosage group displayed the highest lymphocyte proliferation and antibody levels (P<0.05). The median dosage group had the greatest relative body weight gain (89.7%), and the greatest oocyst shedding reduction (53.7%). These results indicate that median dosage of DNA vaccine had good immunogenicity and immune protection effects, and may be used in field applications for coccidiosis control.
Collapse
Affiliation(s)
- Jinjun Xu
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China
| | | | | |
Collapse
|
6
|
Zhao Y, Tahiliani V, Salek-Ardakani S, Croft M. Targeting 4-1BB (CD137) to enhance CD8 T cell responses with poxviruses and viral antigens. Front Immunol 2012; 3:332. [PMID: 23162550 PMCID: PMC3492829 DOI: 10.3389/fimmu.2012.00332] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 10/19/2012] [Indexed: 11/13/2022] Open
Abstract
Attenuated vaccinia virus (VACV) vectors are considered prime vaccine candidates for use in immunotherapy of infectious disease. In spite of this, recent data show that the level of attenuation may hamper the efficient generation of protective CD8 T cells. This suggests that additional adjuvant-like activities may need to be combined with attenuated VACV for optimal vaccination. Stimulatory reagents to the TNFR family molecule 4-1BB (CD137) may represent such an adjuvant for vaccination. Previous murine studies have found that 4-1BB can participate in optimal priming of effector and memory CD8 T cells in response to several virus infections, and concordantly direct stimulation of 4-1BB with agonist reagents effectively boosts the CD8 T cell response against those viruses. In contrast, we recently reported that 4-1BB plays no role in the response to a virulent strain of VACV, questioning whether agonists of 4-1BB will be useful adjuvants for vaccination with VACV vectors. Here we show that agonist anti-4-1BB strongly enhanced the primary viral-specific effector CD8 T cell response during infection with live virulent VACV and attenuated VACV, and during immunization with VACV peptides given in IFA. However, accumulation of memory CD8 T cells was enhanced only following infection with virulent VACV or with peptide vaccination, but not with attenuated VACV, correlating in part with more transient expression of 4-1BB on CD8 T cells with attenuated virus. Our data therefore suggest that 4-1BB may be a promising candidate for targeting as an adjuvant for short-term enhancement of CD8 T cell responses with VACV vaccine strategies, but additional receptors may need to be engaged with 4-1BB to allow long-term CD8 T cell immunity with attenuated VACV vectors.
Collapse
Affiliation(s)
- Yuan Zhao
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology La Jolla, CA, USA
| | | | | | | |
Collapse
|
7
|
Hallengärd D, Bråve A, Isaguliants M, Blomberg P, Enger J, Stout R, King A, Wahren B. A combination of intradermal jet-injection and electroporation overcomes in vivo dose restriction of DNA vaccines. GENETIC VACCINES AND THERAPY 2012; 10:5. [PMID: 22873174 PMCID: PMC3532290 DOI: 10.1186/1479-0556-10-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 07/12/2012] [Indexed: 01/04/2023]
Abstract
Background The use of optimized delivery devices has been shown to enhance the potency of DNA vaccines. However, further optimization of DNA vaccine delivery is needed for this vaccine modality to ultimately be efficacious in humans. Methods Herein we evaluated antigen expression and immunogenicity after intradermal delivery of different doses of DNA vaccines by needle or by the Biojector jet-injection device, with or without the addition of electroporation (EP). Results Neither needle injection augmented by EP nor Biojector alone could induce higher magnitudes of immune responses after immunizations with a high dose of DNA. After division of a defined DNA dose into multiple skin sites, the humoral response was particularly enhanced by Biojector while cellular responses were particularly enhanced by EP. Furthermore, a close correlation between in vivo antigen expression and cell-mediated as well as humoral immune responses was observed. Conclusions These results show that two optimized DNA vaccine delivery devices can act together to overcome dose restrictions of plasmid DNA vaccines.
Collapse
Affiliation(s)
- David Hallengärd
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Nobels väg 16, 171 77, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Scriba TJ, Tameris M, Mansoor N, Smit E, van der Merwe L, Mauff K, Hughes E, Moyo S, Brittain N, Lawrie A, Mulenga H, de Kock M, Gelderbloem S, Veldsman A, Hatherill M, Geldenhuys H, Hill AV, Hussey GD, Mahomed H, Hanekom WA, McShane H. Dose-Finding Study of the Novel Tuberculosis Vaccine, MVA85A, in Healthy BCG-Vaccinated Infants. J Infect Dis 2011; 203:1832-43. [DOI: 10.1093/infdis/jir195] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
9
|
Climent N, Guerra S, García F, Rovira C, Miralles L, Gómez CE, Piqué N, Gil C, Gatell JM, Esteban M, Gallart T. Dendritic cells exposed to MVA-based HIV-1 vaccine induce highly functional HIV-1-specific CD8(+) T cell responses in HIV-1-infected individuals. PLoS One 2011; 6:e19644. [PMID: 21625608 PMCID: PMC3097254 DOI: 10.1371/journal.pone.0019644] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 04/06/2011] [Indexed: 01/04/2023] Open
Abstract
Currently, MVA virus vectors carrying HIV-1 genes are being developed as HIV-1/AIDS prophylactic/therapeutic vaccines. Nevertheless, little is known about the impact of these vectors on human dendritic cells (DC) and their capacity to present HIV-1 antigens to human HIV-specific T cells. This study aimed to characterize the interaction of MVA and MVA expressing the HIV-1 genes Env-Gag-Pol-Nef of clade B (referred to as MVA-B) in human monocyte-derived dendritic cells (MDDC) and the subsequent processes of HIV-1 antigen presentation and activation of memory HIV-1-specific T lymphocytes. For these purposes, we performed ex vivo assays with MDDC and autologous lymphocytes from asymptomatic HIV-infected patients. Infection of MDDC with MVA-B or MVA, at the optimal dose of 0.3 PFU/MDDC, induced by itself a moderate degree of maturation of MDDC, involving secretion of cytokines and chemokines (IL1-ra, IL-7, TNF-α, IL-6, IL-12, IL-15, IL-8, MCP-1, MIP-1α, MIP-1β, RANTES, IP-10, MIG, and IFN-α). MDDC infected with MVA or MVA-B and following a period of 48 h or 72 h of maturation were able to migrate toward CCL19 or CCL21 chemokine gradients. MVA-B infection induced apoptosis of the infected cells and the resulting apoptotic bodies were engulfed by the uninfected MDDC, which cross-presented HIV-1 antigens to autologous CD8+ T lymphocytes. MVA-B-infected MDDC co-cultured with autologous T lymphocytes induced a highly functional HIV-specific CD8+ T cell response including proliferation, secretion of IFN-γ, IL-2, TNF-α, MIP-1β, MIP-1α, RANTES and IL-6, and strong cytotoxic activity against autologous HIV-1-infected CD4+ T lymphocytes. These results evidence the adjuvant role of the vector itself (MVA) and support the clinical development of prophylactic and therapeutic anti-HIV vaccines based on MVA-B.
Collapse
Affiliation(s)
- Núria Climent
- Service of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Raska M, Takahashi K, Czernekova L, Zachova K, Hall S, Moldoveanu Z, Elliott MC, Wilson L, Brown R, Jancova D, Barnes S, Vrbkova J, Tomana M, Smith PD, Mestecky J, Renfrow MB, Novak J. Glycosylation patterns of HIV-1 gp120 depend on the type of expressing cells and affect antibody recognition. J Biol Chem 2010; 285:20860-9. [PMID: 20439465 PMCID: PMC2898351 DOI: 10.1074/jbc.m109.085472] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 04/28/2010] [Indexed: 01/18/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) entry is mediated by the interaction between a variably glycosylated envelope glycoprotein (gp120) and host-cell receptors. Approximately half of the molecular mass of gp120 is contributed by N-glycans, which serve as potential epitopes and may shield gp120 from immune recognition. The role of gp120 glycans in the host immune response to HIV-1 has not been comprehensively studied at the molecular level. We developed a new approach to characterize cell-specific gp120 glycosylation, the regulation of glycosylation, and the effect of variable glycosylation on antibody reactivity. A model oligomeric gp120 was expressed in different cell types, including cell lines that represent host-infected cells or cells used to produce gp120 for vaccination purposes. N-Glycosylation of gp120 varied, depending on the cell type used for its expression and the metabolic manipulation during expression. The resultant glycosylation included changes in the ratio of high-mannose to complex N-glycans, terminal decoration, and branching. Differential glycosylation of gp120 affected envelope recognition by polyclonal antibodies from the sera of HIV-1-infected subjects. These results indicate that gp120 glycans contribute to antibody reactivity and should be considered in HIV-1 vaccine design.
Collapse
Affiliation(s)
- Milan Raska
- From the Departments of Immunology and
- the Departments of Microbiology
| | | | | | | | | | | | | | | | | | | | | | - Jana Vrbkova
- Mathematical Analysis and Applications of Mathematics, Palacky University in Olomouc, Olomouc 77100, Czech Republic
| | | | - Phillip D. Smith
- Medicine, and
- the Veterans Affairs Medical Center, Birmingham, Alabama 35205, and
| | - Jiri Mestecky
- the Departments of Microbiology
- Medicine, and
- the Institute of Microbiology and Immunology, First Faculty of Medicine, Charles University, Prague, Czech Republic 12108
| | - Matthew B. Renfrow
- Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | | |
Collapse
|
11
|
Nájera JL, Gómez CE, García-Arriaza J, Sorzano CO, Esteban M. Insertion of vaccinia virus C7L host range gene into NYVAC-B genome potentiates immune responses against HIV-1 antigens. PLoS One 2010; 5:e11406. [PMID: 20613977 PMCID: PMC2894869 DOI: 10.1371/journal.pone.0011406] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Accepted: 06/08/2010] [Indexed: 11/18/2022] Open
Abstract
Background The highly attenuated vaccinia virus strain NYVAC expressing HIV-1 components has been evaluated as a vaccine candidate in preclinical and clinical trials with encouraging results. We have previously described that the presence of C7L in the NYVAC genome prevents the induction of apoptosis and renders the vector capable of replication in human and murine cell lines while maintaining an attenuated phenotype in mice. Methodology/Principal Findings In an effort to improve the immunogenicity of NYVAC, we have developed a novel poxvirus vector by inserting the VACV host-range C7L gene into the genome of NYVAC-B, a recombinant virus that expresses four HIV-1 antigens from clade B (Env, Gag, Pol and Nef) (referred as NYVAC-B-C7L). In the present study, we have compared the in vitro and in vivo behavior of NYVAC-B and NYVAC-B-C7L. In cultured cells, NYVAC-B-C7L expresses higher levels of heterologous antigen than NYVAC-B as determined by Western blot and fluorescent-activated cell sorting to score Gag expressing cells. In a DNA prime/poxvirus boost approach with BALB/c mice, both recombinants elicited robust, broad and multifunctional antigen-specific T-cell responses to the HIV-1 immunogens expressed from the vectors. However, the use of NYVAC-B-C7L as booster significantly enhanced the magnitude of the T cell responses, and induced a more balanced cellular immune response to the HIV-1 antigens in comparison to that elicited in animals boosted with NYVAC-B. Conclusions/Significance These findings demonstrate the possibility to enhance the immunogenicity of the highly attenuated NYVAC vector by the insertion of the host-range gene C7L and suggest the use of this modified vector as an improved vaccine candidate against HIV/AIDS.
Collapse
Affiliation(s)
- José Luis Nájera
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Carlos Oscar Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
- * E-mail:
| |
Collapse
|
12
|
Immune response induced by a linear DNA vector: influence of dose, formulation and route of injection. Vaccine 2010; 28:3642-9. [PMID: 20362204 DOI: 10.1016/j.vaccine.2010.03.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 03/09/2010] [Accepted: 03/17/2010] [Indexed: 11/26/2022]
Abstract
Previously, minimalistic, immunogenetically defined gene expression (MIDGE) vectors were developed as effective and sophisticated carriers for DNA vaccination. Here we evaluate the influence of dose, formulation and delivery route on the immune response after vaccination with MIDGE-Th1 vectors encoding hepatitis B virus surface antigen (HBsAg). An HBsAg-specific IgG1 and IgG2a antibody response was induced in a dose-dependent manner, whereas the IgG2a/IgG1 ratio was independent of the injected DNA dose. Formulation of MIDGE-HBsAg-Th1 with the cationic pyridinium amphiphile SAINT-18 significantly increased antibody levels of IgG1 and IgG2a compared to the unformulated vector. In contrast, SAINT-18 had neither a significant effect on the IgG2a/IgG1 ratio nor on the type and strength of cellular immunity. Overall, the strongest immune response was generated after intradermal injection, followed by intramuscular and subcutaneous (s.c.) injection. The results show that the formulation of MIDGE-Th1 with SAINT-18 increased the efficacy of the MIDGE-Th1 DNA vaccine and is therefore a suitable approach to improve the efficacy of DNA vaccines also in large animals and humans.
Collapse
|
13
|
Brown SA, Surman SL, Sealy R, Jones BG, Slobod KS, Branum K, Lockey TD, Howlett N, Freiden P, Flynn P, Hurwitz JL. Heterologous Prime-Boost HIV-1 Vaccination Regimens in Pre-Clinical and Clinical Trials. Viruses 2010; 2:435-467. [PMID: 20407589 PMCID: PMC2855973 DOI: 10.3390/v2020435] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/12/2010] [Accepted: 01/22/2010] [Indexed: 12/21/2022] Open
Abstract
Currently, there are more than 30 million people infected with HIV-1 and thousands more are infected each day. Vaccination is the single most effective mechanism for prevention of viral disease, and after more than 25 years of research, one vaccine has shown somewhat encouraging results in an advanced clinical efficacy trial. A modified intent-to-treat analysis of trial results showed that infection was approximately 30% lower in the vaccine group compared to the placebo group. The vaccine was administered using a heterologous prime-boost regimen in which both target antigens and delivery vehicles were changed during the course of inoculations. Here we examine the complexity of heterologous prime-boost immunizations. We show that the use of different delivery vehicles in prime and boost inoculations can help to avert the inhibitory effects caused by vector-specific immune responses. We also show that the introduction of new antigens into boost inoculations can be advantageous, demonstrating that the effect of `original antigenic sin' is not absolute. Pre-clinical and clinical studies are reviewed, including our own work with a three-vector vaccination regimen using recombinant DNA, virus (Sendai virus or vaccinia virus) and protein. Promising preliminary results suggest that the heterologous prime-boost strategy may possibly provide a foundation for the future prevention of HIV-1 infections in humans.
Collapse
Affiliation(s)
- Scott A. Brown
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (S.A.B.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Sherri L. Surman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Robert Sealy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Bart G. Jones
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Karen S. Slobod
- Early Development, Novartis Vaccines and Diagnostics, 350 Mass Ave. Cambridge, MA 02139, USA; E-Mail: (K.S.S.)
| | - Kristen Branum
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Timothy D. Lockey
- Department of Therapeutics, Production and Quality, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (T.D.L.)
| | - Nanna Howlett
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Pamela Freiden
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Patricia Flynn
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
- Department of Pediatrics, University of Tennessee, Memphis, TN 38163, USA
| | - Julia L. Hurwitz
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (S.A.B.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
- Department of Pathology, University of Tennessee, Memphis, TN 38163, USA
| |
Collapse
|
14
|
Scriba TJ, Tameris M, Mansoor N, Smit E, van der Merwe L, Isaacs F, Keyser A, Moyo S, Brittain N, Lawrie A, Gelderbloem S, Veldsman A, Hatherill M, Hawkridge A, Hill AV, Hussey GD, Mahomed H, McShane H, Hanekom WA. Modified vaccinia Ankara-expressing Ag85A, a novel tuberculosis vaccine, is safe in adolescents and children, and induces polyfunctional CD4+ T cells. Eur J Immunol 2010; 40:279-90. [PMID: 20017188 PMCID: PMC3044835 DOI: 10.1002/eji.200939754] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Modified vaccinia Ankara-expressing Ag85A (MVA85A) is a new tuberculosis (TB) vaccine aimed at enhancing immunity induced by BCG. We investigated the safety and immunogenicity of MVA85A in healthy adolescents and children from a TB endemic region, who received BCG at birth. Twelve adolescents and 24 children were vaccinated and followed up for 12 or 6 months, respectively. Adverse events were documented and vaccine-induced immune responses assessed by IFN-gamma ELISpot and intracellular cytokine staining. The vaccine was well tolerated and there were no vaccine-related serious adverse events. MVA85A induced potent and durable T-cell responses. Multiple CD4+ T-cell subsets, based on expression of IFN-gamma, TNF-alpha, IL-2, IL-17 and GM-CSF, were induced. Polyfunctional CD4+ T cells co-expressing IFN-gamma, TNF-alpha and IL-2 dominated the response in both age groups. A novel CD4+ cell subset co-expressing these three Th1 cytokines and IL-17 was induced in adolescents, while a novel CD4+ T-cell subset co-expressing Th1 cytokines and GM-CSF was induced in children. Ag-specific CD8+ T cells were not detected. We conclude that in adolescents and children MVA85A safely induces the type of immunity thought to be important in protection against TB. This includes induction of novel Th1-cell populations that have not been previously described in humans.
Collapse
Affiliation(s)
- Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Michele Tameris
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Nazma Mansoor
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Erica Smit
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Linda van der Merwe
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Fatima Isaacs
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Alana Keyser
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Sizulu Moyo
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Nathaniel Brittain
- Centre for Clinical Vaccinology and Tropical Medicine & The Jenner Institute Laboratories, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Alison Lawrie
- Centre for Clinical Vaccinology and Tropical Medicine & The Jenner Institute Laboratories, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Sebastian Gelderbloem
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
- Aeras Global Tuberculosis Vaccine Foundation, Rondebosch, South Africa
| | - Ashley Veldsman
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Anthony Hawkridge
- Aeras Global Tuberculosis Vaccine Foundation, Rondebosch, South Africa
| | - Adrian V.S. Hill
- Centre for Clinical Vaccinology and Tropical Medicine & The Jenner Institute Laboratories, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Gregory D. Hussey
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Hassan Mahomed
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| | - Helen McShane
- Centre for Clinical Vaccinology and Tropical Medicine & The Jenner Institute Laboratories, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Willem A. Hanekom
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Observatory, South Africa
| |
Collapse
|
15
|
Characterization of T-cell responses in macaques immunized with a single dose of HIV DNA vaccine. J Virol 2009; 84:1243-53. [PMID: 19923181 DOI: 10.1128/jvi.01846-09] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The optimization of immune responses (IR) induced by HIV DNA vaccines in humans is one of the great challenges in the development of an effective vaccine against AIDS. Ideally, this vaccine should be delivered in a single dose to immunize humans. We recently demonstrated that the immunization of mice with a single dose of a DNA vaccine derived from pathogenic SHIV(KU2) (Delta4SHIV(KU2)) induced long-lasting, potent, and polyfunctional HIV-specific CD8(+) T-cell responses (G. Arrode, R. Hegde, A. Mani, Y. Jin, Y. Chebloune, and O. Narayan, J. Immunol. 178:2318-2327, 2007). In the present work, we expanded the characterization of the IR induced by this DNA immunization protocol to rhesus macaques. Animals immunized with a single high dose of Delta4SHIV(KU2) DNA vaccine were monitored longitudinally for vaccine-induced IR using multiparametric flow cytometry-based assays. Interestingly, all five immunized macaques developed broad and polyfunctional HIV-specific T-cell IR that persisted for months, with an unusual reemergence in the blood following an initial decline but in the absence of antibody responses. The majority of vaccine-specific CD4(+) and CD8(+) T cells lacked gamma interferon production but showed high antigen-specific proliferation capacities. Proliferative CD8(+) T cells expressed the lytic molecule granzyme B. No integrated viral vector could be detected in mononuclear cells from immunized animals, and this high dose of DNA did not induce any detectable autoimmune responses against DNA. Taken together, our comprehensive analysis demonstrated for the first time the capacity of a single high dose of HIV DNA vaccine alone to induce long-lasting and polyfunctional T-cell responses in the nonhuman primate model, bringing new insights for the design of future HIV vaccines.
Collapse
|
16
|
Arefian E, Bamdad T, Soleimanjahi H, Akhoond MR, Parsania M, Ghaemi A. A kinetic study of gamma interferon production in herpes simplex virus-1 DNA prime-protein boost regimen comparing to DNA or subunit vaccination. Mol Biol 2009. [DOI: 10.1134/s0026893309030066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Cristillo AD, Galmin L, Restrepo S, Hudacik L, Suschak J, Lewis B, Draghia-Akli R, Aziz N, Weiss D, Markham P, Pal R. HIV-1 Env vaccine comprised of electroporated DNA and protein co-administered with Talabostat. Biochem Biophys Res Commun 2008; 370:22-6. [PMID: 18329382 DOI: 10.1016/j.bbrc.2008.02.145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 02/29/2008] [Indexed: 11/26/2022]
Abstract
Selection of potent yet low reactogenic adjuvants for protein immunization is important for HIV-1 vaccine development. Immunogenicity of electroporated DNA (HIV env) and recombinant gp120, administered with either QS-21 or the orally administered immunomodulator, Talabostat, was evaluated in BALB/c mice. Electroporation of low dose DNA elicited Th1 cytokines and anti-envelope antibodies. Immunization with gp120 protein alone with or without Talabostat elicited lower Th1 and Th2 cytokine levels but comparable anti-gp120 antibodies to QS-21-formulated protein. Boosting of DNA-primed mice with gp120/Talabostat induced similar anti-gp120 antibody titers and slightly higher levels of Th1 and Th2 cytokines relative to QS-21-formulated protein. Induction of CD8(+) and CD4(+) T cells and functional CTL activity was noted. These results highlight the potential use of orally administered Talabostat for efficient protein boosting of antibody and T-cell responses primed by DNA.
Collapse
Affiliation(s)
- Anthony D Cristillo
- Advanced BioScience Laboratories, Inc., 5510 Nicholson Lane, Kensington, MD 20895, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Pillai VB, Hellerstein M, Yu T, Amara RR, Robinson HL. Comparative studies on in vitro expression and in vivo immunogenicity of supercoiled and open circular forms of plasmid DNA vaccines. Vaccine 2008; 26:1136-41. [PMID: 18242791 PMCID: PMC2692637 DOI: 10.1016/j.vaccine.2007.10.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 09/20/2007] [Accepted: 10/01/2007] [Indexed: 11/20/2022]
Abstract
Here we use tests for in vitro expression and in vivo immunogenicty to compare the biological activity of supercoiled and open circular forms of plasmid DNA vaccines. The different forms of vaccine DNA revealed no differences in the expression of mRNA or protein following DEAE-dextran-assisted transfection of cultured cells. In contrast, following intramuscular saline injections in mice, supercoiled DNA was three times more effective than open circular DNA at priming a MVA-boosted CD8 T cell response. Thus, under our experimental conditions, measurements for supercoiled vaccine DNA provided a more accurate assessment of the potential to prime a CD8 response than tests for expression in transiently transfected cells.
Collapse
Affiliation(s)
- Vinod Bhaskara Pillai
- Yerkes National Primate Research Center and Emory Vaccine Center of Emory University School of Medicine, 954 Gatewood Road, Atlanta, GA 30329, USA.
| | | | | | | | | |
Collapse
|
19
|
Cristillo AD, Weiss D, Hudacik L, Restrepo S, Galmin L, Suschak J, Draghia-Akli R, Markham P, Pal R. Persistent antibody and T cell responses induced by HIV-1 DNA vaccine delivered by electroporation. Biochem Biophys Res Commun 2008; 366:29-35. [DOI: 10.1016/j.bbrc.2007.11.052] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 11/10/2007] [Indexed: 10/22/2022]
|
20
|
Wyatt LS, Earl PL, Vogt J, Eller LA, Chandran D, Liu J, Robinson HL, Moss B. Correlation of immunogenicities and in vitro expression levels of recombinant modified vaccinia virus Ankara HIV vaccines. Vaccine 2008; 26:486-93. [PMID: 18155813 PMCID: PMC2262837 DOI: 10.1016/j.vaccine.2007.11.036] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Revised: 11/05/2007] [Accepted: 11/15/2007] [Indexed: 11/23/2022]
Abstract
The purpose of the present study was to correlate the in vitro level of HIV Env expression by recombinant modified vaccinia virus Ankara (rMVA) with immunogenicity in mice. A 5-fold difference in Env synthesis was achieved at the translational level by the presence or absence of an out-of-frame initiation codon upstream of the env gene. This perturbation had no effect on the size or processing of Env. In contrast to the variation in Env synthesis, the rMVAs produced similar amounts of HIV Gag, which were expressed from identical cassettes. Mice immunized with the higher Env expressing rMVAs had about 15-fold higher titers of Env antibodies and several fold higher frequencies of Env-specific CD8+ and CD4+ T cells than mice immunized with the low expresser. The greater immune response achieved by high expression was maintained over a 100-fold dose range. Importantly, enhanced Env immune responses did not come at the expense of lower Gag T cell responses. These data suggest that for high immunogenicity, rMVAs should be engineered to produce the most recombinant protein that can be achieved without compromising the growth and stability of the rMVA.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- Antibody Specificity
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/biosynthesis
- Gene Expression
- Genetic Vectors
- HIV Antibodies/blood
- HIV Antibodies/immunology
- HIV Infections/immunology
- Humans
- Injections, Intramuscular
- Mice
- Mice, Inbred BALB C
- Recombination, Genetic
- Spleen/immunology
- Vaccination
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccinia virus
- env Gene Products, Human Immunodeficiency Virus/genetics
- env Gene Products, Human Immunodeficiency Virus/immunology
- gag Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Linda S Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892-3210, USA.
| | | | | | | | | | | | | | | |
Collapse
|