1
|
Bollimpelli VS, Reddy PBJ, Gangadhara S, Charles TP, Burton SL, Tharp GK, Styles TM, Labranche CC, Smith JC, Upadhyay AA, Sahoo A, Legere T, Shiferaw A, Velu V, Yu T, Tomai M, Vasilakos J, Kasturi SP, Shaw GM, Montefiori D, Bosinger SE, Kozlowski PA, Pulendran B, Derdeyn CA, Hunter E, Amara RR. Intradermal but not intramuscular modified vaccinia Ankara immunizations protect against intravaginal tier2 simian-human immunodeficiency virus challenges in female macaques. Nat Commun 2023; 14:4789. [PMID: 37553348 PMCID: PMC10409804 DOI: 10.1038/s41467-023-40430-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
Route of immunization can markedly influence the quality of immune response. Here, we show that intradermal (ID) but not intramuscular (IM) modified vaccinia Ankara (MVA) vaccinations provide protection from acquisition of intravaginal tier2 simian-human immunodeficiency virus (SHIV) challenges in female macaques. Both routes of vaccination induce comparable levels of serum IgG with neutralizing and non-neutralizing activities. The protection in MVA-ID group correlates positively with serum neutralizing and antibody-dependent phagocytic activities, and envelope-specific vaginal IgA; while the limited protection in MVA-IM group correlates only with serum neutralizing activity. MVA-ID immunizations induce greater germinal center Tfh and B cell responses, reduced the ratio of Th1 to Tfh cells in blood and showed lower activation of intermediate monocytes and inflammasome compared to MVA-IM immunizations. This lower innate activation correlates negatively with induction of Tfh responses. These data demonstrate that the MVA-ID vaccinations protect against intravaginal SHIV challenges by modulating the innate and T helper responses.
Collapse
Affiliation(s)
- Venkata S Bollimpelli
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Pradeep B J Reddy
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Sailaja Gangadhara
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Tysheena P Charles
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Samantha L Burton
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Gregory K Tharp
- NHP Genomics Core Laboratory, Emory National Primate Research Center, Atlanta, GA, 30329, USA
| | - Tiffany M Styles
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Celia C Labranche
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Justin C Smith
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Amit A Upadhyay
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Anusmita Sahoo
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Traci Legere
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Ayalnesh Shiferaw
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Vijayakumar Velu
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Tianwei Yu
- Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Mark Tomai
- 3M Corporate Research and Materials Lab, Saint Paul, MN, USA
| | | | - Sudhir P Kasturi
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - George M Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Steven E Bosinger
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Bali Pulendran
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Cynthia A Derdeyn
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Eric Hunter
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Rama R Amara
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA.
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
2
|
Cho M, Min X, Son HS. Analysis of evolutionary and genetic patterns in structural genes of primate lentiviruses. Genes Genomics 2022; 44:773-791. [PMID: 35511321 PMCID: PMC9068864 DOI: 10.1007/s13258-022-01257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/09/2022] [Indexed: 12/01/2022]
Abstract
Background Primate lentiviruses (HIV1, HIV2, and Simian immunodeficiency virus [SIV]) cause immune deficiency, encephalitis, and infectious anemia in mammals such as cattle, cat, goat, sheep, horse, and puma. Objective This study was designed and conducted with the main purpose of confirming the overall codon usage pattern of primate lentiviruses and exploring the evolutionary and genetic characteristics commonly or specifically expressed in HIV1, HIV2, and SIV. Methods The gag, pol, and env gene sequences of HIV1, HIV2, and SIV were analyzed to determine their evolutionary relationships, nucleotide compositions, codon usage patterns, neutrality, selection pressure (influence of mutational pressure and natural selection), and viral adaptation to human codon usage. Results A strong ‘A’ bias was confirmed in all three structural genes, consistent with previous findings regarding HIV. Notably, the ENC-GC3s plot and neutral evolution analysis showed that all primate lentiviruses were more affected by selection pressure than by mutation caused by the GC composition of the gene, consistent with prior reports regarding HIV1. The overall codon usage bias of pol was highest among the structural genes, while the codon usage bias of env was lowest. The virus groups showing high codon bias in all three genes were HIV1 and SIVcolobus. The codon adaptation index (CAI) and similarity D(A, B) values indicated that although there was a high degree of similarity to human codon usage in all three structural genes of HIV, this similarity was not caused by translation pressure. In addition, compared with HIV1, the codon usage of HIV2 is more similar to the human codon usage, but the overall codon usage bias is lower. Conclusion The origin viruses of HIV (SIVcpz_gor and SIVsmm) exhibit greater similarity to human codon usage in the gag gene, confirming their robust adaptability to human codon usage. Therefore, HIV1 and HIV2 may have evolved to avoid human codon usage by selection pressure in the gag gene after interspecies transmission from SIV hosts to humans. By overcoming safety and stability issues, information from codon usage analysis will be useful for attenuated HIV1 vaccine development. A recoded HIV1 variant can be used as a vaccine vector or in immunotherapy to induce specific innate immune responses. Further research regarding HIV1 dinucleotide usage and codon pair usage will facilitate new approaches to the treatment of AIDS.
Collapse
Affiliation(s)
- Myeongji Cho
- Laboratory of Computational Virology & Viroinformatics, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.,Institute of Health and Environment, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Xianglan Min
- Laboratory of Computational Virology & Viroinformatics, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Hyeon S Son
- Laboratory of Computational Virology & Viroinformatics, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea. .,Institute of Health and Environment, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea. .,Interdisciplinary Graduate Program in Bioinformatics, College of Natural Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
3
|
Ghattas M, Dwivedi G, Lavertu M, Alameh MG. Vaccine Technologies and Platforms for Infectious Diseases: Current Progress, Challenges, and Opportunities. Vaccines (Basel) 2021; 9:1490. [PMID: 34960236 PMCID: PMC8708925 DOI: 10.3390/vaccines9121490] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/11/2021] [Indexed: 01/09/2023] Open
Abstract
Vaccination is a key component of public health policy with demonstrated cost-effective benefits in protecting both human and animal populations. Vaccines can be manufactured under multiple forms including, inactivated (killed), toxoid, live attenuated, Virus-like Particles, synthetic peptide, polysaccharide, polysaccharide conjugate (glycoconjugate), viral vectored (vector-based), nucleic acids (DNA and mRNA) and bacterial vector/synthetic antigen presenting cells. Several processes are used in the manufacturing of vaccines and recent developments in medical/biomedical engineering, biology, immunology, and vaccinology have led to the emergence of innovative nucleic acid vaccines, a novel category added to conventional and subunit vaccines. In this review, we have summarized recent advances in vaccine technologies and platforms focusing on their mechanisms of action, advantages, and possible drawbacks.
Collapse
Affiliation(s)
- Majed Ghattas
- Department of Chemical Engineering, Polytechnique Montreal, Montreal, QC H3T 1J4, Canada;
- Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC H3T 1J4, Canada
| | - Garima Dwivedi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Marc Lavertu
- Department of Chemical Engineering, Polytechnique Montreal, Montreal, QC H3T 1J4, Canada;
- Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC H3T 1J4, Canada
| | - Mohamad-Gabriel Alameh
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- AexeRNA Therapeutics, Washington, DC 20001, USA
| |
Collapse
|
4
|
Human Immunodeficiency Virus C.1086 Envelope gp140 Protein Boosts following DNA/Modified Vaccinia Virus Ankara Vaccination Fail To Enhance Heterologous Anti-V1V2 Antibody Response and Protection against Clade C Simian-Human Immunodeficiency Virus Challenge. J Virol 2019; 93:JVI.00934-19. [PMID: 31341049 DOI: 10.1128/jvi.00934-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/17/2019] [Indexed: 12/29/2022] Open
Abstract
The RV144 human immunodeficiency virus type 1 (HIV-1) vaccine trial showed a strong association between anti-gp70 V1V2 scaffold (V1V2) and anti-V2 hot spot peptide (V2 HS) antibody responses and reduced risk of HIV infection. Accordingly, a primary goal for HIV vaccines is to enhance the magnitude and breadth of V1V2 and V2 HS antibody responses in addition to neutralizing antibodies. Here, we tested the immunogenicity and efficacy of HIV-1 C.1086 gp140 boosts administered sequentially after priming with CD40L-adjuvanted DNA/simian-human immunodeficiency virus (SHIV) and boosting with modified vaccinia virus Ankara (MVA)-SHIV vaccines in rhesus macaques. The DNA/MVA vaccination induced robust vaccine-specific CD4 and CD8 T cell responses with a polyfunctional profile. Two gp140 booster immunizations induced very high levels (∼2 mg/ml) of gp140 binding antibodies in serum, with strong reactivity directed against the homologous (C.1086) V1V2, V2 HS, V3, and gp41 immunodominant (ID) proteins. However, the vaccine-induced antibody showed 10-fold (peak) and 32-fold (prechallenge) weaker binding to the challenge virus (SHIV1157ipd3N4) V1V2 and failed to bind to the challenge virus V2 HS due to a single amino acid change. Point mutations in the immunogen V2 HS to match the V2 HS in the challenge virus significantly diminished the binding of vaccine-elicited antibodies to membrane-anchored gp160. Both vaccines failed to protect from infection following repeated SHIV1157ipd3N4 intrarectal challenges. However, only the protein-boosted animals showed enhanced viral control. These results demonstrate that C.1086 gp140 protein immunizations administered following DNA/MVA vaccination do not significantly boost heterologous V1V2 and V2 HS responses and fail to enhance protection against heterologous SHIV challenge.IMPORTANCE HIV, the virus that causes AIDS, is responsible for millions of infections and deaths annually. Despite intense research for the past 25 years, there remains no safe and effective vaccine available. The significance of this work is in identifying the pros and cons of adding a protein boost to an already well-established DNA/MVA HIV vaccine that is currently being tested in the clinic. Characterizing the effects of the protein boost can allow researchers going forward to design vaccines that generate responses that will be more effective against HIV. Our results in rhesus macaques show that boosting with a specific HIV envelope protein does not significantly boost antibody responses that were identified as immune correlates of protection in a moderately successful RV144 HIV vaccine trial in humans and highlight the need for the development of improved HIV envelope immunogens.
Collapse
|
5
|
Clade C HIV-1 Envelope Vaccination Regimens Differ in Their Ability To Elicit Antibodies with Moderate Neutralization Breadth against Genetically Diverse Tier 2 HIV-1 Envelope Variants. J Virol 2019; 93:JVI.01846-18. [PMID: 30651354 DOI: 10.1128/jvi.01846-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/03/2019] [Indexed: 01/09/2023] Open
Abstract
The goals of preclinical HIV vaccine studies in nonhuman primates are to develop and test different approaches for their ability to generate protective immunity. Here, we compared the impact of 7 different vaccine modalities, all expressing the HIV-1 1086.C clade C envelope (Env), on (i) the magnitude and durability of antigen-specific serum antibody responses and (ii) autologous and heterologous neutralizing antibody capacity. These vaccination regimens included immunization with different combinations of DNA, modified vaccinia virus Ankara (MVA), soluble gp140 protein, and different adjuvants. Serum samples collected from 130 immunized monkeys at two key time points were analyzed using the TZM-bl cell assay: at 2 weeks after the final immunization (week 40/41) and on the day of challenge (week 58). Key initial findings were that inclusion of a gp140 protein boost had a significant impact on the magnitude and durability of Env-specific IgG antibodies, and addition of 3M-052 adjuvant was associated with better neutralizing activity against the SHIV1157ipd3N4 challenge virus and a heterologous HIV-1 CRF01 Env, CNE8. We measured neutralization against a panel of 12 tier 2 Envs using a newly described computational tool to quantify serum neutralization potency by factoring in the predetermined neutralization tier of each reference Env. This analysis revealed modest neutralization breadth, with DNA/MVA immunization followed by gp140 protein boosts in 3M-052 adjuvant producing the best scores. This study highlights that protein-containing regimens provide a solid foundation for the further development of novel adjuvants and inclusion of trimeric Env immunogens that could eventually elicit a higher level of neutralizing antibody breadth.IMPORTANCE Despite much progress, we still do not have a clear understanding of how to elicit a protective neutralizing antibody response against HIV-1 through vaccination. There have been great strides in the development of envelope immunogens that mimic the virus particle, but less is known about how different vaccination modalities and adjuvants contribute to shaping the antibody response. We compared seven different vaccines that were administered to rhesus macaques and that delivered the same envelope protein through various modalities and with different adjuvants. The results demonstrate that some vaccine components are better than others at eliciting neutralizing antibodies with breadth.
Collapse
|
6
|
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
|
7
|
HIV-1 gp120 and Modified Vaccinia Virus Ankara (MVA) gp140 Boost Immunogens Increase Immunogenicity of a DNA/MVA HIV-1 Vaccine. J Virol 2017; 91:JVI.01077-17. [PMID: 29021394 PMCID: PMC5709589 DOI: 10.1128/jvi.01077-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/07/2017] [Indexed: 11/20/2022] Open
Abstract
An important goal of human immunodeficiency virus (HIV) vaccine design is identification of strategies that elicit effective antiviral humoral immunity. One novel approach comprises priming with DNA and boosting with modified vaccinia virus Ankara (MVA) expressing HIV-1 Env on virus-like particles. In this study, we evaluated whether the addition of a gp120 protein in alum or MVA-expressed secreted gp140 (MVAgp140) could improve immunogenicity of a DNA prime-MVA boost vaccine. Five rhesus macaques per group received two DNA primes at weeks 0 and 8 followed by three MVA boosts (with or without additional protein or MVAgp140) at weeks 18, 26, and 40. Both boost immunogens enhanced the breadth of HIV-1 gp120 and V1V2 responses, antibody-dependent cellular cytotoxicity (ADCC), and low-titer tier 1B and tier 2 neutralizing antibody responses. However, there were differences in antibody kinetics, linear epitope specificity, and CD4 T cell responses between the groups. The gp120 protein boost elicited earlier and higher peak responses, whereas the MVAgp140 boost resulted in improved antibody durability and comparable peak responses after the final immunization. Linear V3 specific IgG responses were particularly enhanced by the gp120 boost, whereas the MVAgp140 boost also enhanced responses to linear C5 and C2.2 epitopes. Interestingly, gp120, but not the MVAgp140 boost, increased peak CD4+ T cell responses. Thus, both gp120 and MVAgp140 can augment potential protection of a DNA/MVA vaccine by enhancing gp120 and V1/V2 antibody responses, whereas potential protection by gp120, but not MVAgp140 boosts, may be further impacted by increased CD4+ T cell responses. IMPORTANCE Prior immune correlate analyses with humans and nonhuman primates revealed the importance of antibody responses in preventing HIV-1 infection. A DNA prime-modified vaccinia virus Ankara (MVA) boost vaccine has proven to be potent in eliciting antibody responses. Here we explore the ability of boosts with recombinant gp120 protein or MVA-expressed gp140 to enhance antibody responses elicited by the GOVX-B11 DNA prime-MVA boost vaccine. We found that both types of immunogen boosts enhanced potentially protective antibody responses, whereas the gp120 protein boosts also increased CD4+ T cell responses. Our data provide important information for HIV vaccine designs that aim for effective and balanced humoral and T cell responses.
Collapse
|
8
|
HIV transmitted/founder vaccines elicit autologous tier 2 neutralizing antibodies for the CD4 binding site. PLoS One 2017; 12:e0177863. [PMID: 29020058 PMCID: PMC5636061 DOI: 10.1371/journal.pone.0177863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/04/2017] [Indexed: 01/16/2023] Open
Abstract
Here we report the construction, antigenicity and initial immunogenicity testing of DNA and modified vaccinia Ankara (MVA) vaccines expressing virus-like particles (VLPs) displaying sequential clade C Envelopes (Envs) that co-evolved with the elicitation of broadly neutralizing antibodies (bnAbs) to the CD4 binding site (CD4bs) in HIV-infected individual CH0505. The VLP-displayed Envs showed reactivity for conformational epitopes displayed on the receptor-binding form of Env. Two inoculations of the DNA-T/F vaccine, followed by 3 inoculations of the MVA-T/F vaccine and a final inoculation of the MVA-T/F plus a gp120-T/F protein vaccine elicited nAb to the T/F virus in 2 of 4 rhesus macaques (ID50 of ~175 and ~30). Neutralizing Ab plateaued at 100% neutralization and mapped to the CD4bs like the bnAbs elicited in CH0505. The nAb did not have breadth for other tier 2 viruses. Immunizations with T/F followed by directed-lineage vaccines, both with and without co-delivery of directed-lineage gp120 boosts, failed to elicit tier 2 neutralizing Ab for the CD4bs. Thus, pulsed exposures to DNA and MVA-expressed VLPs plus gp120 protein of a T/F Env can induce autologous tier 2 nAbs to the CD4bs.
Collapse
|
9
|
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
|
10
|
Buchbinder SP, Grunenberg NA, Sanchez BJ, Seaton KE, Ferrari G, Moody MA, Frahm N, Montefiori DC, Hay CM, Goepfert PA, Baden LR, Robinson HL, Yu X, Gilbert PB, McElrath MJ, Huang Y, Tomaras GD. Immunogenicity of a novel Clade B HIV-1 vaccine combination: Results of phase 1 randomized placebo controlled trial of an HIV-1 GM-CSF-expressing DNA prime with a modified vaccinia Ankara vaccine boost in healthy HIV-1 uninfected adults. PLoS One 2017; 12:e0179597. [PMID: 28727817 PMCID: PMC5519050 DOI: 10.1371/journal.pone.0179597] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 05/30/2017] [Indexed: 12/20/2022] Open
Abstract
Background A phase 1 trial of a clade B HIV vaccine in HIV-uninfected adults evaluated the safety and immunogenicity of a DNA prime co-expressing GM-CSF (Dg) followed by different numbers and intervals of modified vaccinia Ankara Boosts (M). Both vaccines produce virus-like particles presenting membrane-bound Env. Methods Four US sites randomized 48 participants to receiving 1/10th the DNA dose as DgDgMMM given at 0, 2, 4, 6 and 8 months, or full dose DgDgM_M or DgDgMM_M regimens, given at 0, 2, 4, and 8 months, and 0, 2, 4, 6, and 10 months, respectively. Peak immunogenicity was measured 2 weeks post-last vaccination. Results All regimens were well tolerated and safe. Full dose DgDgM_M and DgDgMM_M regimens generated Env-specific IgG to HIV-1 Env in >90%, IgG3 in >80%, and IgA in <20% of participants. Responses to gp140 and gp41 targets were more common and of higher magnitude than to gp120 and V1V2. The gp41 antibody included reactivity to the conserved immunodominant region with specificities known to mediate virus capture and phagocytosis and did not cross-react with a panel of intestinal flora antigens. The 3rd dose of MVA increased the avidity of elicited antibody (7.5% to 39%), the ADCC response to Bal gp120 (14% to 64%), and the one-year durability of the IgG3 responses to gp41 by 4-fold (13% vs. 3.5% retention of peak response). The co-expressed GM-CSF did not enhance responses over those in trials testing this vaccine without GM-CSF. Conclusion This DNA/MVA prime-boost regimen induced durable, functional humoral responses that included ADCC, high antibody avidity, and Env IgG1 and IgG3 binding responses to the immunodominant region of gp41. The third, spaced MVA boost improved the overall quality of the antibody response. These products without co-expressed GM-CSF but combined with protein boosts will be considered for efficacy evaluation. Trial registration ClinicalTrials.gov NCT01571960
Collapse
Affiliation(s)
- Susan P. Buchbinder
- Bridge HIV, San Francisco Department of Public Health, San Francisco, California, United States of America
- Departments of Medicine, Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
- * E-mail:
| | - Nicole A. Grunenberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Brittany J. Sanchez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kelly E. Seaton
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Guido Ferrari
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - M. Anthony Moody
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Christine M. Hay
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Paul A. Goepfert
- Department of Medicine, University of Alabama, Birmingham, Alabama, United States of America
| | - Lindsey R. Baden
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | | | - Xuesong Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Georgia D. Tomaras
- Department of Surgery, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | | |
Collapse
|
11
|
Chapman R, Jongwe TI, Douglass N, Chege G, Williamson AL. Heterologous prime-boost vaccination with DNA and MVA vaccines, expressing HIV-1 subtype C mosaic Gag virus-like particles, is highly immunogenic in mice. PLoS One 2017; 12:e0173352. [PMID: 28278263 PMCID: PMC5344398 DOI: 10.1371/journal.pone.0173352] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/19/2017] [Indexed: 12/31/2022] Open
Abstract
In an effort to make affordable vaccines suitable for the regions most affected by HIV-1, we have constructed stable vaccines that express an HIV-1 subtype C mosaic Gag immunogen (BCG-GagM, MVA-GagM and DNA-GagM). Mosaic immunogens have been designed to address the tremendous diversity of this virus. Here we have shown that GagM buds from cells infected and transfected with MVA-GagM and DNA-GagM respectively and forms virus-like particles. Previously we showed that a BCG-GagM prime MVA-GagM boost generated strong cellular immune responses in mice. In this study immune responses to the DNA-GagM and MVA-GagM vaccines were evaluated in homologous and heterologous prime-boost vaccinations. The DNA homologous prime boost vaccination elicited predominantly CD8+ T cells while the homologous MVA vaccination induced predominantly CD4+ T cells. A heterologous DNA-GagM prime MVA-GagM boost induced strong, more balanced Gag CD8+ and CD4+ T cell responses and that were predominantly of an effector memory phenotype. The immunogenicity of the mosaic Gag (GagM) was compared to a naturally occurring subtype C Gag (GagN) using a DNA homologous vaccination regimen. DNA-GagN expresses a natural Gag with a sequence that was closest to the consensus sequence of subtype C viruses sampled in South Africa. DNA-GagM homologous vaccination induced cumulative HIV-1 Gag-specific IFN-γ ELISPOT responses that were 6.5-fold higher than those induced by the DNA-GagN vaccination. Similarly, DNA-GagM vaccination generated 7-fold higher levels of cytokine-positive CD8+ T cells than DNA-GagN, indicating that this subtype C mosaic Gag elicits far more potent immune responses than a consensus-type Gag. Cells transfected and infected with DNA-GagM and MVA-GagM respectively, expressed high levels of GagM and produced budding virus-like particles. Our data indicates that a heterologous prime boost regimen using DNA and MVA vaccines expressing HIV-1 subtype C mosaic Gag is highly immunogenic in mice and warrants further investigation in non-human primates.
Collapse
Affiliation(s)
- Ros Chapman
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Tsungai Ivai Jongwe
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicola Douglass
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gerald Chege
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
12
|
Thompson M, Heath SL, Sweeton B, Williams K, Cunningham P, Keele BF, Sen S, Palmer BE, Chomont N, Xu Y, Basu R, Hellerstein MS, Kwa S, Robinson HL. DNA/MVA Vaccination of HIV-1 Infected Participants with Viral Suppression on Antiretroviral Therapy, followed by Treatment Interruption: Elicitation of Immune Responses without Control of Re-Emergent Virus. PLoS One 2016; 11:e0163164. [PMID: 27711228 PMCID: PMC5053438 DOI: 10.1371/journal.pone.0163164] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/14/2016] [Indexed: 12/01/2022] Open
Abstract
GV-TH-01, a Phase 1 open-label trial of a DNA prime—Modified Vaccinia Ankara (MVA) boost vaccine (GOVX-B11), was undertaken in HIV infected participants on antiretroviral treatment (ART) to evaluate safety and vaccine-elicited T cell responses, and explore the ability of elicited CD8+ T cells to control viral rebound during analytical treatment interruption (TI). Nine men who began antiretroviral therapy (ART) within 18 months of seroconversion and had sustained plasma HIV-1 RNA <50 copies/mL for at least 6 months were enrolled. Median age was 38 years, median pre-ART HIV-1 RNA was 140,000 copies/ml and mean baseline CD4 count was 755/μl. Two DNA, followed by 2 MVA, inoculations were given 8 weeks apart. Eight subjects completed all vaccinations and TI. Clinical and laboratory adverse events were generally mild, with no serious or grade 4 events. Only reactogenicity events were considered related to study drug. No treatment emergent viral resistance was seen. The vaccinations did not reduce viral reservoirs and virus re-emerged in all participants during TI, with a median time to re-emergence of 4 weeks. Eight of 9 participants had CD8+ T cells that could be stimulated by vaccine-matched Gag peptides prior to vaccination. Vaccinations boosted these responses as well as eliciting previously undetected CD8+ responses. Elicited T cells did not display signs of exhaustion. During TI, temporal patterns of viral re-emergence and Gag-specific CD8+ T cell expansion suggested that vaccine-specific CD8+ T cells had been stimulated by re-emergent virus in only 2 of 8 participants. In these 2, transient decreases in viremia were associated with Gag selection in known CD8+ T cell epitopes. We hypothesize that escape mutations, already archived in the viral reservoir, plus a poor ability of CD8+ T cells to traffic to and control virus at sites of re-emergence, limited the therapeutic efficacy of the DNA/MVA vaccine. TRIAL REGISTRATION clinicaltrials.gov NCT01378156.
Collapse
Affiliation(s)
- Melanie Thompson
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Sonya L. Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Bentley Sweeton
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Kathy Williams
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Pamela Cunningham
- Alabama Vaccine Research Clinic, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sharon Sen
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Brent E. Palmer
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Nicolas Chomont
- Centre de recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Canada
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Rahul Basu
- GeoVax, Inc., Atlanta, Georgia, United States of America
| | | | - Suefen Kwa
- GeoVax, Inc., Atlanta, Georgia, United States of America
| | | |
Collapse
|
13
|
Chamcha V, Kannanganat S, Gangadhara S, Nabi R, Kozlowski PA, Montefiori DC, LaBranche CC, Wrammert J, Keele BF, Balachandran H, Sahu S, Lifton M, Santra S, Basu R, Moss B, Robinson HL, Amara RR. Strong, but Age-Dependent, Protection Elicited by a Deoxyribonucleic Acid/Modified Vaccinia Ankara Simian Immunodeficiency Virus Vaccine. Open Forum Infect Dis 2016; 3:ofw034. [PMID: 27006959 PMCID: PMC4800464 DOI: 10.1093/ofid/ofw034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/09/2016] [Indexed: 11/12/2022] Open
Abstract
Background. In this study, we analyzed the protective efficacy of a simian immunodeficiency virus (SIV) macaque 239 (SIVmac239) analogue of the clinically tested GOVX-B11 deoxyribonucleic acid (DNA)/modified vaccinia Ankara (MVA) human immunodeficiency virus vaccine. Methods. The tested vaccine used a DNA immunogen mutated to mimic the human vaccine and a regimen with DNA deliveries at weeks 0 and 8 and MVA deliveries at weeks 16 and 32. Twelve weekly rectal challenges with 0.3 animal infectious doses of SIV sootey mangabey E660 (SIVsmE660) were administered starting at 6 months after the last immunization. Results. Over the first 6 rectal exposures to SIVsmE660, <10-year-old tripartite motif-containing protein 5 (TRIM5)α-permissive rhesus macaques showed an 80% reduction in per-exposure risk of infection as opposed to a 46% reduction in animals over 10 years old; and, over the 12 challenges, they showed a 72% as opposed to a 10% reduction. Analyses of elicited immune responses suggested that higher antibody responses in the younger animals had played a role in protection. Conclusions. The simian analogue of the GOVX-B11 HIV provided strong protection against repeated rectal challenges in young adult macaques.
Collapse
Affiliation(s)
| | - Sunil Kannanganat
- Yerkes National Primate Research Center, Emory University , Atlanta, Georgia
| | - Sailaja Gangadhara
- Yerkes National Primate Research Center, Emory University , Atlanta, Georgia
| | - Rafiq Nabi
- Department of Microbiology , Immunology and Parasitology, Louisiana State University Health Sciences Center , New Orleans
| | - Pamela A Kozlowski
- Department of Microbiology , Immunology and Parasitology, Louisiana State University Health Sciences Center , New Orleans
| | | | | | - Jens Wrammert
- Department of Pediatrics , Emory University School of Medicine , Atlanta, Georgia
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc. , Frederick National Laboratory for Cancer Research , Maryland
| | | | - Sujata Sahu
- Harvard Medical School, Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - Michelle Lifton
- Harvard Medical School, Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - Sampa Santra
- Harvard Medical School, Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | | | - Bernard Moss
- Laboratory of Viral Diseases , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland
| | | | - Rama Rao Amara
- Yerkes National Primate Research Center, Emory University , Atlanta, Georgia
| |
Collapse
|
14
|
Williamson AL, Rybicki EP. Justification for the inclusion of Gag in HIV vaccine candidates. Expert Rev Vaccines 2015; 15:585-98. [PMID: 26645951 DOI: 10.1586/14760584.2016.1129904] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
It is widely accepted that effective human immunodeficiency virus (HIV) vaccines need to elicit a range of responses, including neutralising antibodies and T-cells. In natural HIV infections, immune responses to Gag are associated with lower viral load in infected individuals, and these responses can be measured against infected cells before the replication of HIV. Priming immune responses to Gag with DNA or recombinant Bacillus Calmette-Guérin (BCG) vaccines, and boosting with Gag virus-like particles as subunit vaccines or Gag produced in vivo by other vaccine vectors, elicits high-magnitude, broad polyfunctional responses, with memory T-cell responses appropriate for virus control. This review provides justification for the inclusion of HIV Gag in vaccine regimens, either as a transgene expressing protein that may assemble to form budded particles, or as purified virus-like particles. Possible benefits would include early control via CD8(+) T-cells at the site of infection, control of spread from the entry portal, and control of viraemia if infection is established.
Collapse
Affiliation(s)
- Anna-Lise Williamson
- a Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town , South Africa.,b National Health Laboratory Service, Groote Schuur Hospital, Cape Town and Department of Pathology , University of Cape Town , Cape Town , South Africa
| | - Edward P Rybicki
- a Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town , South Africa.,c Biopharming Research Unit, Department of Molecular and Cell Biology , University of Cape Town , Cape Town , South Africa
| |
Collapse
|
15
|
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
|
16
|
Ramirez LA, Arango T, Boyer J. Therapeutic and prophylactic DNA vaccines for HIV-1. Expert Opin Biol Ther 2015; 13:563-73. [PMID: 23477730 DOI: 10.1517/14712598.2013.758709] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION DNA vaccines have moved into clinical trials in several fields and their success will be important for licensure of this vaccine modality. An effective vaccine for HIV-1 remains elusive and the development of one is troubled by safety and efficacy issues. Additionally, the ability for an HIV-1 vaccine to induce both the cellular and humoral arms of the immune system is needed. DNA vaccines not only offer a safe approach for the development of an HIV-1 vaccine but they have also been shown to elicit both arms of the immune system. AREAS COVERED This review explores how DNA vaccine design including the regimen, genetic adjuvants used, targeting, and mode of delivery continues to undergo improvements, thereby providing a potential option for an immunogenic vaccine for HIV-1. EXPERT OPINION Continued improvements in delivery technology, in particular electroporation, and the use of prime-boost vaccine strategies will aid in boosting the immunogenicity of DNA vaccines. Basic immunology research will also help discover new potential adjuvant targets that can be combined with DNA vaccination, such as inhibitors of inhibitory receptors.
Collapse
Affiliation(s)
- Lorenzo Antonio Ramirez
- University of Pennsylvania, Pathology, Stellar Chance Labs, 422 Curie Blvd, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
17
|
Bet A, Sterret S, Sato A, Bansal A, Goepfert PA. Characterization of T-cell responses to cryptic epitopes in recipients of a noncodon-optimized HIV-1 vaccine. J Acquir Immune Defic Syndr 2014; 65:142-50. [PMID: 24442221 PMCID: PMC3896890 DOI: 10.1097/qai.0b013e3182a9917e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Cryptic epitopes (CEs) can be encoded by any of the 5 alternative reading frames (ARFs, 2 sense and 3 antisense) of a known gene. Although CE responses are commonly detected during HIV-1 infection, it is not known whether these responses are induced after vaccination. METHODS Using a bioinformatic approach, we determined that vaccines with codon-optimized HIV inserts significantly skewed CE sequences and are not likely to induce crossreactive responses to natural HIV CE. We then evaluated the CE- and protein-specific T-cell responses using Gag, Pol, and ARF peptide pools among participants immunized with a non-codon optimized vaccine regimen of 2 pGA2/JS7 DNA primes followed by 2 MVA/HIV62 Gag-Pol-Env vector boosts or 4 saline injections. RESULTS Vaccinees had significantly more interferon gamma enzyme-linked immunosorbent spot (IFNγ ELISpot) responses toward Gag (P = 0.003) but not toward Pol protein than did placebo recipients. However, CE-specific T-cell responses were low in magnitude, and their frequencies did not differ significantly between vaccine and placebo recipients. Additionally, most positive CE responses could not be mapped to individual peptides. After expanding responses in a cultured assay, however, the frequency and the median magnitude of responses to ARF peptides were significantly greater in vaccinees (P < 0.0001), indicating that CE-specific T-cell responses are present but below an ex vivo assay's limit of detection. CONCLUSIONS Our data demonstrate that HIV-1 vaccines currently in clinical trials are poorly immunogenic with regard to CE-specific T-cell responses. Therefore, the context of HIV-1 immunogens may need to be modified as a comprehensive strategy to broaden vaccine-induced T-cell responses.
Collapse
Affiliation(s)
- Anne Bet
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL USA 35294
| | - Sarah Sterret
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL USA 35294
| | - Alicia Sato
- Statistical Center for HIV/AIDS Research & Prevention (SCHARP), Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024
| | - Anju Bansal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA 35294
| | - Paul A. Goepfert
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL USA 35294
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA 35294
| |
Collapse
|
18
|
Scotti N, Rybicki EP. Virus-like particles produced in plants as potential vaccines. Expert Rev Vaccines 2014; 12:211-24. [DOI: 10.1586/erv.12.147] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
19
|
Goepfert PA, Elizaga ML, Seaton K, Tomaras GD, Montefiori DC, Sato A, Hural J, DeRosa SC, Kalams SA, McElrath MJ, Keefer MC, Baden LR, Lama JR, Sanchez J, Mulligan MJ, Buchbinder SP, Hammer SM, Koblin BA, Pensiero M, Butler C, Moss B, Robinson HL. Specificity and 6-month durability of immune responses induced by DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J Infect Dis 2014; 210:99-110. [PMID: 24403557 DOI: 10.1093/infdis/jiu003] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Clade B DNA and recombinant modified vaccinia Ankara (MVA) vaccines producing virus-like particles displaying trimeric membrane-bound envelope glycoprotein (Env) were tested in a phase 2a trial in human immunodeficiency virus (HIV)-uninfected adults for safety, immunogenicity, and 6-month durability of immune responses. METHODS A total of 299 individuals received 2 doses of JS7 DNA vaccine and 2 doses of MVA/HIV62B at 0, 2, 4, and 6 months, respectively (the DDMM regimen); 3 doses of MVA/HIV62B at 0, 2, and 6 months (the MMM regimen); or placebo injections. RESULTS At peak response, 93.2% of the DDMM group and 98.4% of the MMM group had binding antibodies for Env. These binding antibodies were more frequent and of higher magnitude for the transmembrane subunit (gp41) than the receptor-binding subunit (gp120) of Env. For both regimens, response rates were higher for CD4(+) T cells (66.4% in the DDMM group and 43.1% in the MMM group) than for CD8(+) T cells (21.8% in the DDMM group and 14.9% in the MMM group). Responding CD4(+) and CD8(+) T cells were biased toward Gag, and >70% produced 2 or 3 of the 4 cytokines evaluated (ie, interferon γ, interleukin 2, tumor necrosis factor α, and granzyme B). Six months after vaccination, the magnitudes of antibodies and T-cell responses had decreased by <3-fold. CONCLUSIONS DDMM and MMM vaccinations with virus-like particle-expressing immunogens elicited durable antibody and T-cell responses.
Collapse
Affiliation(s)
| | - Marnie L Elizaga
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Kelly Seaton
- Laboratory for AIDS Vaccine Research and Development, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Georgia D Tomaras
- Laboratory for AIDS Vaccine Research and Development, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - David C Montefiori
- Laboratory for AIDS Vaccine Research and Development, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Alicia Sato
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Stephen C DeRosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center University of Washington, Seattle, Washington
| | - Spyros A Kalams
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center University of Washington, Seattle, Washington
| | - Michael C Keefer
- University of Rochester School of Medicine and Dentistry, Rochester
| | - Lindsey R Baden
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Javier R Lama
- Asociacion Civil IMPACTA Salud y Educacion, Lima, Peru
| | - Jorge Sanchez
- Asociacion Civil IMPACTA Salud y Educacion, Lima, Peru
| | | | | | | | | | | | | | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | | | | |
Collapse
|
20
|
Lindsay RW, Ouellette I, Arendt HE, Martinez J, DeStefano J, Lopez M, Pavlakis GN, Chiuchiolo MJ, Parks CL, King CR. SIV antigen-specific effects on immune responses induced by vaccination with DNA electroporation and plasmid IL-12. Vaccine 2013; 31:4749-58. [PMID: 23954384 DOI: 10.1016/j.vaccine.2013.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/28/2013] [Accepted: 08/05/2013] [Indexed: 10/26/2022]
Abstract
Molecular adjuvants are important for augmenting or modulating immune responses induced by DNA vaccination. Promising results have been obtained using IL-12 expression plasmids in a variety of disease models including the SIV model of HIV infection. We used a mouse model to evaluate plasmid IL-12 (pIL-12) in a DNA prime, recombinant adenovirus serotype 5 (rAd5) boost regimen specifically to evaluate the effect of IL-12 expression on cellular and humoral immunity induced against both SIVmac239 Gag and Env antigens. Priming with electroporated (EP) DNA+pIL-12 resulted in a 2-4-fold enhanced frequency of Gag-specific CD4 T cells which was maintained through the end of the study irrespective of the pIL-12 dose, while memory Env-specific CD4+T cells were maintained only at the low dose of pIL-12. There was little positive effect of pIL-12 on the humoral response to Env, and in fact, high dose pIL-12 dramatically reduced SIV Env-specific IgG. Additionally, both doses of pIL-12 diminished the frequency of CD8 T-cells after DNA prime, although a rAd5 boost recovered CD8 responses regardless of the pIL-12 dose. In this prime-boost regimen, we have shown that a high dose pIL-12 can systemically reduce Env-specific humoral responses and CD4T cell frequency, but not Gag-specific CD4+ T cells. These data indicate that it is important to independently characterize individual SIV or HIV antigen immunogenicity in multi-antigenic vaccines as a function of adjuvant dose.
Collapse
Affiliation(s)
- Ross W Lindsay
- International AIDS Vaccine Initiative, 140 58th Street, Brooklyn, NY 11220, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wallace A, West K, Rothman AL, Ennis FA, Lu S, Wang S. Post-translational intracellular trafficking determines the type of immune response elicited by DNA vaccines expressing Gag antigen of Human Immunodeficiency Virus Type 1 (HIV-1). Hum Vaccin Immunother 2013; 9:2095-102. [PMID: 23941868 DOI: 10.4161/hv.26009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In the current study, immune responses induced by Gag DNA vaccines with different designs were evaluated in Balb/C mice. The results demonstrated that the DNA vaccine with the full length wild type gag gene (Wt-Gag) mainly produced Gag antigens intracellularly and induced a higher level of cell-mediated immune (CMI) responses, as measured by IFN-gamma ELISPOT, intracellular cytokine staining (ICS), and cytotoxic T lymphocytes (CTL) assays against a dominant CD8(+) T cell epitope (AMQMLKETI). In contrast, the addition of a tissue plasminogen activator (tPA) leader sequence significantly improved overall Gag protein expression/secretion and Gag-specific antibody responses; however, Gag-specific CMI responses were decreased. The mutation of zinc-finger motif changed Gag protein expression patterns and reduced the ability to generate both CMI and antibody responses against Gag. These findings indicate that the structure and post-translational processing of antigens expressed by DNA vaccines play a critical role in eliciting optimal antibody or CMI responses.
Collapse
Affiliation(s)
- Aaron Wallace
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA
| | - Kim West
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA; Center for Infectious Diseases and Vaccine Research; University of Massachusetts Medical School; Worcester, MA USA
| | - Alan L Rothman
- Center for Infectious Diseases and Vaccine Research; University of Massachusetts Medical School; Worcester, MA USA
| | - Francis A Ennis
- Center for Infectious Diseases and Vaccine Research; University of Massachusetts Medical School; Worcester, MA USA
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA
| |
Collapse
|
22
|
Buonaguro L, Tagliamonte M, Visciano ML, Tornesello ML, Buonaguro FM. Developments in virus-like particle-based vaccines for HIV. Expert Rev Vaccines 2013; 12:119-127. [PMID: 23414404 DOI: 10.1586/erv.12.152] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Virus-like particles (VLPs) hold great promise for the development of effective and affordable vaccines. VLPs, indeed, are suitable for presentation and efficient delivery to antigen-presenting cells of linear as well as conformational antigens. This will ultimately result in a crosspresentation with both MHC class I and II molecules to prime CD4(+) T-helper and CD8(+) cytotoxic T cells. This review describes an update on the development and use of VLPs as vaccine approaches for HIV.
Collapse
Affiliation(s)
- Luigi Buonaguro
- Department of Experimental Oncology, Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori 'Fond Pascale', Via Mariano Semmola 142, 80131 Napoli, Italy
| | | | | | | | | |
Collapse
|
23
|
Hellerstein M, Xu Y, Marino T, Lu S, Yi H, Wright ER, Robinson HL. Co-expression of HIV-1 virus-like particles and granulocyte-macrophage colony stimulating factor by GEO-D03 DNA vaccine. Hum Vaccin Immunother 2012; 8:1654-8. [PMID: 23111169 PMCID: PMC3601140 DOI: 10.4161/hv.21978] [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] [Indexed: 01/09/2023] Open
Abstract
Here, we report on GEO-D03, a DNA vaccine that co-expresses non-infectious HIV-1 virus-like particles (VLPs) and the human cytokine, granulocyte-macrophage colony-stimulating factor (GM-CSF). The virus-like particles display the native gp160 form of the HIV-1 Envelope glycoprotein (Env) and are designed to elicit antibody against the natural form of Env on virus and virus-infected cells. The DNA-expressed HIV Gag, Pol and Env proteins also have the potential to elicit virus-specific CD4 and CD8 T cells. The purpose of the co-expressed GM-CSF is to target a cytokine that recruits, expands and differentiates macrophages and dendritic cells to the site of VLP expression. The GEO-D03 DNA vaccine is currently entered into human trials as a prime for a recombinant modified vaccinia Ankara (MVA) boost. In preclinical studies in macaques using an SIV prototype vaccine, this vaccination regimen elicited both anti-viral T cells and antibody, and provided 70% protection against acquisition during 12 weekly rectal exposures with a heterologous SIV. Higher avidity of the Env-specific Ab for the native form of the Env in the challenge virus correlated with lower likelihood of SIV infection.
Collapse
|
24
|
Abstract
The complex interplay between the host immune response and HIV has been the subject of intense research over the last 25 years. HIV and simian immunodeficiency virus (SIV) CD8 T cells have been of particular interest since they were demonstrated to be temporally associated with reduction in virus load shortly following transmission. Here, we briefly review the phenotypic and functional properties of HIV-specific and SIV-specific CD8 T-cell subsets during HIV infection and consider the influence of viral variation with specific responses that are associated with disease progression or control. The development of an effective HIV/AIDS vaccine combined with existing successful prevention and treatment strategies is essential for preventing new infections. In the context of previous clinical HIV/AIDS vaccine trials, we consider the challenges faced by therapeutic and vaccine strategies designed to elicit effective HIV-specific CD8 T cells.
Collapse
|
25
|
Goepfert PA, Elizaga ML, Sato A, Qin L, Cardinali M, Hay CM, Hural J, DeRosa SC, DeFawe OD, Tomaras GD, Montefiori DC, Xu Y, Lai L, Kalams SA, Baden LR, Frey SE, Blattner WA, Wyatt LS, Moss B, Robinson HL. Phase 1 safety and immunogenicity testing of DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J Infect Dis 2011; 203:610-9. [PMID: 21282192 DOI: 10.1093/infdis/jiq105] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recombinant DNA and modified vaccinia virus Ankara (rMVA) vaccines represent a promising approach to an HIV/AIDS vaccine. This Phase 1 clinical trial compared the safety and immunogenicity of a rMVA vaccine administered with and without DNA vaccine priming METHODS GeoVax pGA2/JS7 DNA (D) and MVA/HIV62 (M) vaccines encode noninfectious virus-like particles. Intramuscular needle injections were used to deliver placebo, 2 doses of DNA followed by 2 doses of rMVA (DDMM), one dose of DNA followed by 2 doses of rMVA (DMM), or 3 doses of rMVA (MMM) to HIV-seronegative participants. RESULTS Local and systemic symptoms were mild or moderate. Immune response rates for CD4 + and CD8 + T cells were highest in the DDMM group and lowest in the MMM group (77% vs 43% CD4 + and 42% vs 17% CD8 +). In contrast, response rates for Env binding and neutralizing Ab were highest in the MMM group. The DMM group had intermediate response rates. A 1/10th-dose DDMM regimen induced similar T cell but reduced Ab response rates compared with the full-dose DDMM. CONCLUSIONS MVA62 was well tolerated and elicited different patterns of T cell and Ab responses when administered alone or in combination with the JS7 DNA vaccine.
Collapse
Affiliation(s)
- Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, AL 35294, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Schneider-Ohrum K, Ross TM. Virus-Like Particles for Antigen Delivery at Mucosal Surfaces. Curr Top Microbiol Immunol 2011; 354:53-73. [DOI: 10.1007/82_2011_135] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
27
|
Prime‐boost vaccinations using recombinant flavivirus replicon and vaccinia virus vaccines: an ELISPOT analysis. Immunol Cell Biol 2010; 89:426-36. [DOI: 10.1038/icb.2010.99] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Tenbusch M, Grunwald T, Niezold T, Storcksdieck Genannt Bonsmann M, Hannaman D, Norley S, Uberla K. Codon-optimization of the hemagglutinin gene from the novel swine origin H1N1 influenza virus has differential effects on CD4(+) T-cell responses and immune effector mechanisms following DNA electroporation in mice. Vaccine 2010; 28:3273-7. [PMID: 20206668 DOI: 10.1016/j.vaccine.2010.02.090] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 02/17/2010] [Accepted: 02/17/2010] [Indexed: 12/18/2022]
Abstract
DNA electroporation is a powerful vaccine strategy that could be rapidly adapted to address emerging viruses. We therefore compared cellular and humoral immune responses in mice vaccinated with DNA expression plasmids encoding either the wildtype or a codon-optimized sequence of hemagglutinin from the novel swine origin H1N1 influenza virus. While expression of HA from the wildtype sequence was hardly detectable, the H1N1 hemagglutinin was well expressed from the codon-optimized sequence. Despite poor expression of the wildtype sequence, both plasmids induced similar levels of CD4(+) T-cell responses. However, CD8(+) T-cell and antibody responses were substantially higher after immunization with the codon-optimized DNA vaccine. Thus, efficient induction of immune effector mechanisms against HA of the novel H1N1 influenza virus requires codon-optimization of the DNA vaccines. Since DNA vaccines and several viral vector vaccines employ the same cellular RNA-Polymerase II dependent expression pathway, the poor expression levels from wildtype HA sequences might also limit the induction of immune effector mechanisms by such viral vector vaccines.
Collapse
Affiliation(s)
- M Tenbusch
- Department of Molecular and Medical Virology, Ruhr-Universität Bochum, Universitätsstr. 150, 44801 Bochum, Germany.
| | | | | | | | | | | | | |
Collapse
|
29
|
Adjuvantive effects of anti-4-1BB agonist Ab and 4-1BBL DNA for a HIV-1 Gag DNA vaccine: different effects on cellular and humoral immunity. Vaccine 2009; 28:1300-9. [PMID: 19944789 DOI: 10.1016/j.vaccine.2009.11.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 11/05/2009] [Accepted: 11/06/2009] [Indexed: 12/21/2022]
Abstract
Plasmid DNA immunizations induce low levels but a broad spectrum of cellular and humoral immune responses. Here, we investigate the potential of co-stimulation through 4-1BB as an adjuvant for a HIV-1 DNA vaccine in mice. We designed plasmid DNAs expressing either the membrane bound or soluble form of 4-1BBL, and compared with the agonistic anti-4-1BB Ab for their ability to adjuvant the Gag DNA vaccine. Both, anti-4-1BB agonistic Ab as well as 4-1BBL DNA enhanced the Gag-specific cellular immune responses. However, in complete contrast to the agonistic Ab that suppressed humoral immunity to Gag, 4-1BBL DNA adjuvanted vaccines enhanced Gag-specific IgG responses. Importantly, the expression of Gag and 4-1BBL from the same plasmid was critical for the adjuvant activity. Collectively, our data suggest that for a HIV-1 vaccine where both antigen-specific cellular and humoral immunity are desirable, 4-1BBL expressed by a DNA vaccine is a superior adjuvant than anti-4-1BB agonistic Ab.
Collapse
|
30
|
Dobaño C, Sedegah M, Rogers WO, Kumar S, Zheng H, Hoffman SL, Doolan DL. Plasmodium: Mammalian codon optimization of malaria plasmid DNA vaccines enhances antibody responses but not T cell responses nor protective immunity. Exp Parasitol 2009; 122:112-23. [DOI: 10.1016/j.exppara.2009.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 12/27/2008] [Accepted: 02/18/2009] [Indexed: 11/24/2022]
|
31
|
Zhao J, Lai L, Amara RR, Montefiori DC, Villinger F, Chennareddi L, Wyatt LS, Moss B, Robinson HL. Preclinical studies of human immunodeficiency virus/AIDS vaccines: inverse correlation between avidity of anti-Env antibodies and peak postchallenge viremia. J Virol 2009; 83:4102-11. [PMID: 19224993 PMCID: PMC2668498 DOI: 10.1128/jvi.02173-08] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 02/07/2009] [Indexed: 11/20/2022] Open
Abstract
A major challenge for human immunodeficiency virus (HIV)/AIDS vaccines is the elicitation of anti-Env antibodies (Ab) capable of neutralizing the diversity of isolates in the pandemic. Here, we show that high-avidity, but nonneutralizing, Abs can have an inverse correlation with peak postchallenge viremia for a heterologous challenge. Vaccine studies were conducted in rhesus macaques using DNA priming followed by modified vaccinia Ankara boosting with HIV type 1 (HIV-1) immunogens that express virus-like particles displaying CCR5-tropic clade B (strain ADA) or clade C (IN98012) Envs. Rhesus granulocyte-macrophage colony-stimulating factor was used as an adjuvant for enhancing the avidity of anti-Env Ab responses. Challenge was with simian/human immunodeficiency virus (SHIV)-162P3, a CCR5-tropic clade B chimera of SIV and HIV-1. Within the groups receiving the clade B vaccine, a strong inverse correlation was found between the avidity of anti-Env Abs and peak postchallenge viremia. This correlation required the use of native but not gp120 or gp140 forms of Env for avidity assays. The high-avidity Ab elicited by the ADA Env had excellent breadth for the Envs of incident clade B but not clade C isolates, whereas the high-avidity Ab elicited by the IN98012 Env had excellent breadth for incident clade C but not clade B isolates. High-avidity Ab elicited by a SHIV vaccine with a dual-tropic clade B Env (89.6) had limited breadth for incident isolates. Our results suggest that certain Envs can elicit nonneutralizing but high-avidity Ab with broad potential for blunting incident infections of the same clade.
Collapse
Affiliation(s)
- Jun Zhao
- Emory Vaccine Center, Atlanta, Georgia 30329, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
HIV vaccine research is at a crossroads carefully contemplating on the next path. The unexpected results of the Merck vaccine trial, while providing a stunning blow to a field in dire need of a protective vaccine, has also raised several fundamental questions regarding the candidate immunogen itself, preexisting immunity to vaccine vectors, surrogate assays and animal models used for assessing preclinical protective responses, as well as relevant endpoints to be measured in a clinical trial. As a result, the research community is faced with the daunting task of identifying novel vaccine concepts and products to continue the search. This review highlights and addresses some of the scientific and practical concerns.
Collapse
|
33
|
Huang X, Lu B, Yu W, Fang Q, Liu L, Zhuang K, Shen T, Wang H, Tian P, Zhang L, Chen Z. A novel replication-competent vaccinia vector MVTT is superior to MVA for inducing high levels of neutralizing antibody via mucosal vaccination. PLoS One 2009; 4:e4180. [PMID: 19159014 PMCID: PMC2613559 DOI: 10.1371/journal.pone.0004180] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 12/06/2008] [Indexed: 11/25/2022] Open
Abstract
Mucosal vaccination offers great advantage for inducing protective immune response to prevent viral transmission and dissemination. Here, we report our findings of a head-to-head comparison of two viral vectors modified vaccinia Ankara (MVA) and a novel replication-competent modified vaccinia Tian Tan (MVTT) for inducing neutralizing antibodies (Nabs) via intramuscular and mucosal vaccinations in mice. MVTT is an attenuated variant of the wild-type VTT, which was historically used as a smallpox vaccine for millions of Chinese people. The spike glycoprotein (S) of SARS-CoV was used as the test antigen after the S gene was constructed in the identical genomic location of two vectors to generate vaccine candidates MVTT-S and MVA-S. Using identical doses, MVTT-S induced lower levels (∼2-3-fold) of anti- SARS-CoV neutralizing antibodies (Nabs) than MVA-S through intramuscular inoculation. MVTT-S, however, was capable of inducing consistently 20-to-100-fold higher levels of Nabs than MVA-S when inoculated via either intranasal or intraoral routes. These levels of MVTT-S-induced Nab responses were substantially (∼10-fold) higher than that induced via the intramuscular route in the same experiments. Moreover, pre-exposure to the wild-type VTT via intranasal or intraoral route impaired the Nab response via the same routes of MVTT-S vaccination probably due to the pre-existing anti-VTT Nab response. The efficacy of intranasal or intraoral vaccination, however, was still 20-to-50-fold better than intramuscular inoculation despite the subcutaneous pre-exposure to wild-type VTT. Our data have implications for people who maintain low levels of anti-VTT Nabs after historical smallpox vaccination. MVTT is therefore an attractive live viral vector for mucosal vaccination.
Collapse
Affiliation(s)
- Xiaoxing Huang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
| | - Bin Lu
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
| | - Wenbo Yu
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Qing Fang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
| | - Li Liu
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Ke Zhuang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
| | - Tingting Shen
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
| | - Haibo Wang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Po Tian
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei, People's Republic of China
| | - Linqi Zhang
- AIDS Research Center, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
- Comprehensive AIDS Research Center, Tsinghua University, Beijing, People's Republic of China
| | - Zhiwei Chen
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
- * E-mail:
| |
Collapse
|
34
|
Abstract
Since the discovery, over a decade and a half ago, that genetically engineered DNA can be delivered in vaccine form and elicit an immune response, there has been much progress in understanding the basic biology of this platform. A large amount of data has been generated in preclinical model systems, and more sustained cellular responses and more consistent antibody responses are being observed in the clinic. Four DNA vaccine products have recently been approved, all in the area of veterinary medicine. These results suggest a productive future for this technology as more optimized constructs, better trial designs and improved platforms are being brought into the clinic.
Collapse
Affiliation(s)
- Michele A Kutzler
- Division of Infectious Diseases and HIV Medicine, The Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, USA
| | | |
Collapse
|
35
|
Chege GK, Shephard EG, Meyers A, van Harmelen J, Williamson C, Lynch A, Gray CM, Rybicki EP, Williamson AL. HIV-1 subtype C Pr55gag virus-like particle vaccine efficiently boosts baboons primed with a matched DNA vaccine. J Gen Virol 2008; 89:2214-2227. [PMID: 18753231 DOI: 10.1099/vir.0.83501-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A DNA vaccine expressing human immunodeficiency virus type 1 (HIV-1) southern African subtype C Gag (pTHGag) and a recombinant baculovirus Pr55gag virus-like particle prepared using a subtype C Pr55gag protein (Gag VLP) was tested in a prime-boost inoculation regimen in Chacma baboons. The response of five baboons to Gag peptides in a gamma interferon (IFN-gamma) enzyme-linked immunospot (ELISPOT) assay after three pTHGag immunizations ranged from 100 to 515 spot-forming units (s.f.u.) per 10(6) peripheral blood mononuclear cells (PBMCs), whilst the response of two baboons to the Gag VLP vaccine ranged from 415 to 465 s.f.u. per 10(6) PBMCs. An increase in the Gag-specific response to a range of 775-3583 s.f.u. per 10(6) PBMCs was achieved by boosting with Gag VLPs the five baboons that were primed with pTHGag. No improvement in Gag responses was achieved in this prime-boost inoculation regimen by increasing the number of pTHGag inoculations to six. IFN-gamma responses were mapped to several peptides, some of which have been reported to be targeted by PBMCs from HIV-1 subtype C-infected individuals. Gag VLPs, given as a single-modality regimen, induced a predominantly CD8+ T-cell IFN-gamma response and interleukin-2 was a major cytokine within a mix of predominantly Th1 cytokines produced by a DNA-VLP prime-boost modality. The prime-boost inoculation regimen induced high serum p24 antibody titres in all baboons, which were several fold above that induced by the individual vaccines. Overall, this study demonstrated that these DNA prime/VLP boost vaccine regimens are highly immunogenic in baboons, inducing high-magnitude and broad multifunctional responses, providing support for the development of these products for clinical trials.
Collapse
Affiliation(s)
- Gerald K Chege
- Institute of Primate Research, PO Box 24481, Karen 00502, Nairobi, Kenya.,Medical Virology, Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Enid G Shephard
- MRC/UCT Liver Research Centre, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Ann Meyers
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Rondebosch, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Joanne van Harmelen
- Medical Virology, Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Carolyn Williamson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa.,Medical Virology, Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Alisson Lynch
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Clive M Gray
- National Institute for Communicable Diseases, Private Bag X4, Sandringham 2131, Johannesburg, South Africa
| | - Edward P Rybicki
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Rondebosch, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Anna-Lise Williamson
- National Health Laboratory Service, Groote Schuur Hospital, Observatory, Cape Town, South Africa.,Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa.,Medical Virology, Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, Rondebosch, Cape Town, South Africa
| |
Collapse
|
36
|
Meyers A, Chakauya E, Shephard E, Tanzer FL, Maclean J, Lynch A, Williamson AL, Rybicki EP. Expression of HIV-1 antigens in plants as potential subunit vaccines. BMC Biotechnol 2008; 8:53. [PMID: 18573204 PMCID: PMC2443125 DOI: 10.1186/1472-6750-8-53] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 06/23/2008] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) has infected more than 40 million people worldwide, mainly in sub-Saharan Africa. The high prevalence of HIV-1 subtype C in southern Africa necessitates the development of cheap, effective vaccines. One means of production is the use of plants, for which a number of different techniques have been successfully developed. HIV-1 Pr55Gag is a promising HIV-1 vaccine candidate: we compared the expression of this and a truncated Gag (p17/p24) and the p24 capsid subunit in Nicotiana spp. using transgenic plants and transient expression via Agrobacterium tumefaciens and recombinant tobamovirus vectors. We also investigated the influence of subcellular localisation of recombinant protein to the chloroplast and the endoplasmic reticulum (ER) on protein yield. We partially purified a selected vaccine candidate and tested its stimulation of a humoral and cellular immune response in mice. RESULTS Both transient and transgenic expression of the HIV antigens were successful, although expression of Pr55Gag was low in all systems; however, the Agrobacterium-mediated transient expression of p24 and p17/p24 yielded best, to more than 1 mg p24/kg fresh weight. Chloroplast targeted protein levels were highest in transient and transgenic expression of p24 and p17/p24. The transiently-expressed p17/p24 was not immunogenic in mice as a homologous vaccine, but it significantly boosted a humoral and T cell immune response primed by a gag DNA vaccine, pTHGagC. CONCLUSION Transient agroinfiltration was best for expression of all of the recombinant proteins tested, and p24 and p17/p24 were expressed at much higher levels than Pr55Gag. Our results highlight the usefulness of plastid signal peptides in enhancing the production of recombinant proteins meant for use as vaccines. The p17/p24 protein effectively boosted T cell and humoral responses in mice primed by the DNA vaccine pTHGagC, showing that this plant-produced protein has potential for use as a vaccine.
Collapse
MESH Headings
- AIDS Vaccines/biosynthesis
- AIDS Vaccines/genetics
- Adjuvants, Immunologic/genetics
- Agrobacterium tumefaciens/genetics
- Animals
- Chloroplasts/genetics
- Chloroplasts/metabolism
- Endoplasmic Reticulum/genetics
- Endoplasmic Reticulum/metabolism
- Female
- Gene Expression
- Gene Expression Regulation, Plant
- Genes, gag
- Genetic Vectors
- HIV Antigens/biosynthesis
- HIV Antigens/genetics
- HIV Antigens/immunology
- HIV Infections/immunology
- HIV Seronegativity
- HIV-1/genetics
- Humans
- Mice
- Mice, Inbred BALB C
- Plants, Genetically Modified
- Nicotiana/genetics
- Tobamovirus/genetics
- Transformation, Genetic
- Vaccines, Subunit/biosynthesis
- Vaccines, Subunit/genetics
- gag Gene Products, Human Immunodeficiency Virus/biosynthesis
- gag Gene Products, Human Immunodeficiency Virus/genetics
Collapse
Affiliation(s)
- Ann Meyers
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Ereck Chakauya
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
- CSIR Biosciences, Pretoria 0001, South Africa
| | - Enid Shephard
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- MRC/UCT Liver Research Centre, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Fiona L Tanzer
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - James Maclean
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Alisson Lynch
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Observatory 7925, South Africa
| | - Edward P Rybicki
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| |
Collapse
|
37
|
Ngumbela KC, Ryan KP, Sivamurthy R, Brockman MA, Gandhi RT, Bhardwaj N, Kavanagh DG. Quantitative effect of suboptimal codon usage on translational efficiency of mRNA encoding HIV-1 gag in intact T cells. PLoS One 2008; 3:e2356. [PMID: 18523584 PMCID: PMC2387063 DOI: 10.1371/journal.pone.0002356] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 05/03/2008] [Indexed: 11/18/2022] Open
Abstract
Background The sequences of wild-isolate strains of Human Immunodeficiency Virus-1 (HIV-1) are characterized by low GC content and suboptimal codon usage. Codon optimization of DNA vectors can enhance protein expression both by enhancing translational efficiency, and by altering RNA stability and export. Although gag codon optimization is widely used in DNA vectors and experimental vaccines, the actual effect of altered codon usage on gag translational efficiency has not been quantified. Methodology and Principal Findings To quantify translational efficiency of gag mRNA in live T cells, we transfected Jurkat cells with increasing doses of capped, polyadenylated synthetic mRNA corresponding to wildtype or codon-optimized gag sequences, measured Gag production by quantitative ELISA and flow cytometry, and estimated the translational efficiency of each transcript as pg of Gag antigen produced per µg of input mRNA. We found that codon optimization yielded a small increase in gag translational efficiency (approximately 1.6 fold). In contrast when cells were transfected with DNA vectors requiring nuclear transcription and processing of gag mRNA, codon optimization resulted in a very large enhancement of Gag production. Conclusions We conclude that suboptimal codon usage by HIV-1 results in only a slight loss of gag translational efficiency per se, with the vast majority of enhancement in protein expression from DNA vectors due to altered processing and export of nuclear RNA.
Collapse
Affiliation(s)
- Kholiswa C. Ngumbela
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa
| | - Kieran P. Ryan
- Partners AIDS Research Center, Massachusetts General Hospital and Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rohini Sivamurthy
- Partners AIDS Research Center, Massachusetts General Hospital and Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mark A. Brockman
- Partners AIDS Research Center, Massachusetts General Hospital and Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rajesh T. Gandhi
- Partners AIDS Research Center, Massachusetts General Hospital and Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nina Bhardwaj
- Partners AIDS Research Center, Massachusetts General Hospital and Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
- New York University School of Medicine, New York, New York, United States of America
| | - Daniel G. Kavanagh
- Partners AIDS Research Center, Massachusetts General Hospital and Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
38
|
Lai L, Vödrös D, Kozlowski PA, Montefiori DC, Wilson RL, Akerstrom VL, Chennareddi L, Yu T, Kannanganat S, Ofielu L, Villinger F, Wyatt LS, Moss B, Amara RR, Robinson HL. GM-CSF DNA: an adjuvant for higher avidity IgG, rectal IgA, and increased protection against the acute phase of a SHIV-89.6P challenge by a DNA/MVA immunodeficiency virus vaccine. Virology 2007; 369:153-67. [PMID: 17698160 PMCID: PMC2215061 DOI: 10.1016/j.virol.2007.07.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 07/03/2007] [Accepted: 07/11/2007] [Indexed: 12/01/2022]
Abstract
Single intradermal or intramuscular inoculations of GM-CSF DNA with the DNA prime for a simian-human immunodeficiency virus (SHIV)-89.6 vaccine, which consists of DNA priming followed by modified vaccinia Ankara (MVA) boosting, increased protection of both the blood and intestines against the acute phase of an intrarectal SHIV-89.6P challenge. GM-CSF appeared to contribute to protection by enhancing two antibody responses: the avidity maturation of anti-Env IgG in blood (p=or<0.01) and the presence of long lasting anti-viral IgA in rectal secretions (p<0.01). The avidity of anti-Env IgG showed strong correlations with protection both pre and post challenge. Animals with the highest avidity anti-Env Ab had 1000-fold reductions in peak viremia over those with the lowest avidity anti-Env Ab. The enhanced IgA response was associated with the best protection, but did not achieve significance.
Collapse
Affiliation(s)
- Lilin Lai
- Yerkes National Primate Research Center, 954 Gatewood Road, Atlanta, GA 30329, USA; Emory Vaccine Center, 954 Gatewood Road, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Robinson HL, Sharma S, Zhao J, Kannanganat S, Lai L, Chennareddi L, Yu T, Montefiori DC, Amara RR, Wyatt LS, Moss B. Immunogenicity in macaques of the clinical product for a clade B DNA/MVA HIV vaccine: elicitation of IFN-gamma, IL-2, and TNF-alpha coproducing CD4 and CD8 T cells. AIDS Res Hum Retroviruses 2007; 23:1555-62. [PMID: 18160013 DOI: 10.1089/aid.2007.0165] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The clinical product for a clade B HIV DNA/MVA vaccine expressing Gag, Pol, and Env has been tested for immunogenicity in macaques. Responding T cells were at the threshold for detection following DNA priming at weeks 0 and 8 but underwent sharp expansions and contractions following MVA boosting at weeks 16 and 24. Both CD4 and CD8 T cell responses had high frequencies of cytokine coproducing cells with >50% of the memory cells coproducing multiple cytokines including IL-2. The highest responses were elicited to Gag, followed by Env and then Pol. In two of six macaques, the vaccine also elicited low levels of neutralizing Ab for easy to neutralize clade B isolates.
Collapse
Affiliation(s)
- Harriet L. Robinson
- Emory University, Yerkes National Primate Research Center, Atlanta, Georgia 30329
- Emory Vaccine Center, Atlanta, Georgia 30322
| | - Sunita Sharma
- Emory University, Yerkes National Primate Research Center, Atlanta, Georgia 30329
- Emory Vaccine Center, Atlanta, Georgia 30322
- CCID/NCHHSTP/DHPSE/LB, Atlanta, Georgia 30329
| | - Jun Zhao
- Emory University, Yerkes National Primate Research Center, Atlanta, Georgia 30329
- Emory Vaccine Center, Atlanta, Georgia 30322
| | - Sunil Kannanganat
- Emory University, Yerkes National Primate Research Center, Atlanta, Georgia 30329
- Emory Vaccine Center, Atlanta, Georgia 30322
| | - Lilin Lai
- Emory University, Yerkes National Primate Research Center, Atlanta, Georgia 30329
- Emory Vaccine Center, Atlanta, Georgia 30322
| | - Lakshmi Chennareddi
- Emory University, Yerkes National Primate Research Center, Atlanta, Georgia 30329
- Emory Vaccine Center, Atlanta, Georgia 30322
| | - Tianwei Yu
- Emory Vaccine Center, Atlanta, Georgia 30322
- Emory University, Rollins School of Public Health, Atlanta, Georgia 30322
| | - David C. Montefiori
- Duke University Medical Center, Department of Surgery, Durham, North Carolina 27710
| | - Rama Rao Amara
- Emory University, Yerkes National Primate Research Center, Atlanta, Georgia 30329
- Emory Vaccine Center, Atlanta, Georgia 30322
| | - Linda S. Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892-3210
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892-3210
| |
Collapse
|
40
|
Comparative studies on in vitro expression and in vivo immunogenicity of supercoiled and open circular forms of plasmid DNA vaccines. Vaccine 2007; 26:1136-41. [PMID: 18242791 DOI: 10.1016/j.vaccine.2007.10.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
|
41
|
Nigam P, Earl PL, Americo JL, Sharma S, Wyatt LS, Edghill-Spano Y, Chennareddi LS, Silvera P, Moss B, Robinson HL, Amara RR. DNA/MVA HIV-1/AIDS vaccine elicits long-lived vaccinia virus-specific immunity and confers protection against a lethal monkeypox challenge. Virology 2007; 366:73-83. [PMID: 17507071 PMCID: PMC2072046 DOI: 10.1016/j.virol.2007.04.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 02/05/2007] [Accepted: 04/09/2007] [Indexed: 11/17/2022]
Abstract
Modified vaccinia Ankara (MVA) is being tested in humans as an alternative to the current smallpox vaccine Dryvax. Here, we compare the magnitude and longevity of protective immune responses elicited by a DNA/MVA HIV-1 vaccine with those elicited by Dryvax using a monkeypox virus/macaque model. The DNA/MVA vaccine elicited similar levels of vaccinia virus (VV)-specific antibody and 5-10-fold lower levels of VV-specific cellular responses than Dryvax. This MVA-elicited cellular and humoral immunity was long-lived. A subset of the DNA/MVA- and Dryvax-vaccinated macaques were subjected to a lethal monkeypox virus challenge at 3 years after vaccination. All of the vaccinated monkeys survived, whereas the unvaccinated controls succumbed to monkeypox. The viral control correlated with early postchallenge levels of monkeypox-specific neutralizing antibody but not with VV-specific cellular immune response. Thus, our results demonstrate the elicitation of long lasting protective immunity for a lethal monkeypox challenge by a DNA/MVA HIV-1 vaccine.
Collapse
Affiliation(s)
- Pragati Nigam
- Emory Vaccine Center, Department of Microbiology and Immunology, and Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, NE, Atlanta, GA 30329, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Young KR, McBurney SP, Karkhanis LU, Ross TM. Virus-like particles: designing an effective AIDS vaccine. Methods 2007; 40:98-117. [PMID: 16997718 DOI: 10.1016/j.ymeth.2006.05.024] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2005] [Accepted: 05/05/2006] [Indexed: 01/10/2023] Open
Abstract
Viruses that infect eukaryotic organisms have the unique characteristic of self-assembling into particles. The mammalian immune system is highly attuned to recognizing and attacking these viral particles following infection. The use of particle-based immunogens, often delivered as live-attenuated viruses, has been an effective vaccination strategy for a variety of viruses. The development of an effective vaccine against the human immunodeficiency virus (HIV) has proven to be a challenge, since HIV infects cells of the immune system causing severe immunodeficiency resulting in the syndrome known as AIDS. In addition, the ability of the virus to adapt to immune pressure and reside in an integrated form in host cells presents hurdles for vaccinologists to overcome. A particle-based vaccine strategy has promise for eliciting high titer, long-lived, immune responses to a diverse number of viral epitopes against different HIV antigens. Live-attenuated viruses are effective at generating both cellular and humoral immune responses. However, while these vaccines stimulate immunity, challenged animals rarely clear the viral infection and the degree of attenuation directly correlates with protection from disease. Further, a live-attenuated vaccine has the potential to revert to a pathogenic form. Alternatively, virus-like particles (VLPs) mimic the viral particle without causing an immunodeficiency disease. VLPs are self-assembling, non-replicating, non-pathogenic particles that are similar in size and conformation to intact virions. A variety of VLPs for lentiviruses are currently in preclinical and clinical trials. This review focuses on our current status of VLP-based AIDS vaccines, regarding issues of purification and immune design for animal and clinical trials.
Collapse
Affiliation(s)
- Kelly R Young
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | |
Collapse
|
43
|
McBurney SP, Young KR, Ross TM. Membrane embedded HIV-1 envelope on the surface of a virus-like particle elicits broader immune responses than soluble envelopes. Virology 2006; 358:334-46. [PMID: 17011011 DOI: 10.1016/j.virol.2006.08.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 08/07/2006] [Accepted: 08/22/2006] [Indexed: 11/24/2022]
Abstract
Virally regulated HIV-1 particles were expressed from DNA plasmids encoding Gag, protease, reverse transcriptase, Vpu, Tat, Rev, and Env. The sequences for integrase, Vpr, Vif, Nef, and the long terminal repeats (LTRs) were deleted. Mutations were engineered into the VLP genome to produce particles deficient in activities associated with viral reverse transcriptase, RNase H, and RNA packaging. Each plasmid efficiently secreted particles from primate cells in vitro and particles were purified from the supernatants and used as immunogens. Mice (BALB/c) were vaccinated intranasally (day 1 and weeks 3 and 6) with purified VLPs and the elicited immunity was compared to particles without Env (Gag(p55)), to soluble monomeric Env(gp120), or to soluble trimerized Env(gp140). Only mice vaccinated with VLPs had robust anti-Env cellular immunity. In contrast, all mice had high titer anti-Env serum antibody (IgG). However, VLP-vaccinated mice had antisera that detected a broader number of linear Env peptides, had anti-Env mucosal IgA and IgG, as well as higher titers of serum neutralizing antibodies. VLPs elicited high titer antibodies that recognized linear regions in V4-C5 and the ectodomain of gp41, but did not recognize V3. These lentiviral VLPs are effective mucosal immunogens that elicit broader immunity against Env determinants in both the systemic and mucosal immune compartments than soluble forms of Env.
Collapse
Affiliation(s)
- Sean P McBurney
- Center for Vaccine Research for Emerging Diseases and Biodefense, University of Pittsburgh School of Medicine, 9047 Biomedical Sciences Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
44
|
Wang S, Taaffe J, Parker C, Solórzano A, Cao H, García-Sastre A, Lu S. Hemagglutinin (HA) proteins from H1 and H3 serotypes of influenza A viruses require different antigen designs for the induction of optimal protective antibody responses as studied by codon-optimized HA DNA vaccines. J Virol 2006; 80:11628-37. [PMID: 16987975 PMCID: PMC1642598 DOI: 10.1128/jvi.01065-06] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Effective antibody responses provide crucial immunity against influenza virus infection. The hemagglutinin (HA) protein is the major target of protective antibody responses induced by viral infection and by vaccination with both inactivated and live-attenuated flu vaccines, but knowledge about the optimal designs of protective HA antigens from different flu serotypes is still limited. In this study, we have significantly improved the immunogenicity of HA-expressing DNA vaccines by using codon-optimized HA sequences for either an H1 serotype (A/NewCal/20/99) or an H3 serotype (A/Panama/2007/99) human influenza A virus and then used these constructs as model antigens to identify the optimal HA antigen designs to elicit high-level protective antibody responses. Two forms of HA antigen, a wild-type, full-length HA and a secreted form with transmembrane (TM) domain-truncated HA, were produced. Both forms of HA DNA vaccines, from either H1 or H3 serotypes, were able to elicit high levels of HA-specific immunoglobulin G responses in immunized rabbits as measured by enzyme-linked immunosorbent assay. Interestingly, the abilities of H1 HA and H3 HA antigens to elicit hemagglutination inhibition (HI) and neutralizing antibody (NAb) responses differ. For the H1 HA antigens, the full-length HA induced significantly higher HI and NAb responses than did the TM-truncated HA. For the H3 HA antigen, both the full-length HA and TM-truncated HA induced high levels of HI and NAb responses. These data indicate that H1 and H3 antigens have different expression requirements for the induction of an optimal protective antibody response and that the structure integrity of HA antigens is critical for eliciting type-specific protective antibody responses. Our findings will have an important impact on future subunit-based flu vaccine development.
Collapse
Affiliation(s)
- Shixia Wang
- Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Lazare Research Building, Worcester, MA 01605-2397, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Mulligan MJ, Russell ND, Celum C, Kahn J, Noonan E, Montefiori DC, Ferrari G, Weinhold KJ, Smith JM, Amara RR, Robinson HL. Excellent safety and tolerability of the human immunodeficiency virus type 1 pGA2/JS2 plasmid DNA priming vector vaccine in HIV type 1 uninfected adults. AIDS Res Hum Retroviruses 2006; 22:678-83. [PMID: 16831092 DOI: 10.1089/aid.2006.22.678] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A vaccine consisting of DNA priming followed by recombinant modified vaccinia Ankara (rMVA) boosting has achieved long-term control of a pathogenic challenge with a chimera of simian and human immunodeficiency viruses (SHIV-89.6P) in rhesus macaques. Based on these results, clade B HIV-1 DNA and rMVA immunogens have been developed for trials in humans. We conducted a first-time in humans phase I safety trial using the pGA2/JS2 (JS2) HIV-1 DNA priming vector expressing Gag, Pol, Env, Tat, Rev, and Vpu. Thirty HIV-uninfected adults were vaccinated with 0.3 or 3 mg of JS2 DNA, or a saline placebo, by intramuscular injection at months 0 and 2. Both doses of DNA were safe and well-tolerated with no differences between the control, 0.3 mg, or 3 mg groups (n = 6, 12, and 12, respectively) through 12 months of postvaccination follow- up. A chromium-release assay using fresh peripheral blood mononuclear cells (PBMCs) and a validated IFN-gamma ELISpot assay with frozen PBMCs failed to detect CD4(+) or CD8(+) HIV-1-specific T cell responses. HIV-specific neutralizing antibodies were also not detected. The vaccine is being further developed as a priming vector for a combined DNA plus rMVA prime/boost HIV vaccination regimen.
Collapse
|
46
|
Du L, He Y, Wang Y, Zhang H, Ma S, Wong CK, Wu SH, Ng F, Huang JD, Yuen KY, Jiang S, Zhou Y, Zheng BJ. Recombinant adeno-associated virus expressing the receptor-binding domain of severe acute respiratory syndrome coronavirus S protein elicits neutralizing antibodies: Implication for developing SARS vaccines. Virology 2006; 353:6-16. [PMID: 16793110 PMCID: PMC7111904 DOI: 10.1016/j.virol.2006.03.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 01/27/2006] [Accepted: 03/31/2006] [Indexed: 12/31/2022]
Abstract
Development of an effective vaccine for severe acute respiratory syndrome (SARS) remains to be a priority to prevent possible re-emergence of SARS coronavirus (SARS-CoV). We previously demonstrated that the receptor-binding domain (RBD) of SARS-CoV S protein is a major target of neutralizing antibodies. This suggests that the RBD may serve as an ideal vaccine candidate. Recombinant adeno-associated virus (rAAV) has been proven to be an effective system for gene delivery and vaccine development. In this study, a novel vaccine against SARS-CoV was developed based on the rAAV delivery system. The gene encoding RBD was cloned into a pAAV-IRES-hrGFP plasmid. The immunogenicity induced by the resulting recombinant RBD-rAAV was evaluated in BALB/c mice. The results demonstrated that (1) a single dose of RBD-rAAV vaccination could induce sufficient neutralizing antibody against SARS-CoV infection; (2) two more repeated doses of the vaccination boosted the neutralizing antibody to about 5 times of the level achieved by a single dose of the immunization and (3) the level of the antibody continued to increase for the entire duration of the experiment of 5.5 months. These results suggested that RBD-rAAV is a promising SARS candidate vaccine.
Collapse
Affiliation(s)
- Lanying Du
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuxian He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- Lindsley F. Kimball Research Institute, The New York Blood Center, New York, NY10021, USA
| | - Yijia Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Haojie Zhang
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Selene Ma
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Charlotte K.L. Wong
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Sharon H.W. Wu
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Fai Ng
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jian-Dong Huang
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, The New York Blood Center, New York, NY10021, USA
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- Corresponding authors. Y. Zhou is to be contacted at fax: +86 10 6381 5259. B.-J. Zheng, fax: +8 52 2855 1241.
| | - Bo-Jian Zheng
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Corresponding authors. Y. Zhou is to be contacted at fax: +86 10 6381 5259. B.-J. Zheng, fax: +8 52 2855 1241.
| |
Collapse
|
47
|
Liu J, Hellerstein M, McDonnel M, Amara RR, Wyatt LS, Moss B, Robinson HL. Dose-response studies for the elicitation of CD8 T cells by a DNA vaccine, used alone or as the prime for a modified vaccinia Ankara boost. Vaccine 2006; 25:2951-8. [PMID: 17360078 DOI: 10.1016/j.vaccine.2006.05.081] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 05/16/2006] [Accepted: 05/22/2006] [Indexed: 10/24/2022]
Abstract
Here, we conduct dose-response studies in mice for the elicitation of CD8 T cells by a DNA vaccine that expresses HIV Gag. For DNA doses ranging from 1 to 100 microg, the studies revealed greater than 10-fold increases in anti-Gag CD8 T cells following a DNA prime or a DNA prime and a constant modified vaccinia Ankara (MVA) boost. These results are in contrast to dose-response studies for MVA vectors expressing Gag, where only 2-3-fold increases in anti-Gag CD8 T cells were elicited by 100-fold increases in dose.
Collapse
Affiliation(s)
- Jinyan Liu
- Yerkes National Primate Research Center and Emory Vaccine Center of Emory University, Atlanta, GA, United States
| | | | | | | | | | | | | |
Collapse
|
48
|
Ellenberger D, Wyatt L, Li B, Buge S, Lanier N, Rodriguez IV, Sariol CA, Martinez M, Monsour M, Vogt J, Smith J, Otten R, Montefiori D, Kraiselburd E, Moss B, Robinson H, McNicholl J, Butera S. Comparative immunogenicity in rhesus monkeys of multi-protein HIV-1 (CRF02_AG) DNA/MVA vaccines expressing mature and immature VLPs. Virology 2005; 340:21-32. [PMID: 16023165 DOI: 10.1016/j.virol.2005.06.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2005] [Revised: 05/13/2005] [Accepted: 06/09/2005] [Indexed: 11/22/2022]
Abstract
We developed an AIDS vaccine for Western and West-Central Africa founded on HIV-1 subtype CRF02_AG. Rhesus macaques were primed with Gag-Pol-Env-expressing plasmid DNA and boosted with a recombinant modified vaccinia virus Ankara (rMVA), expressing matched proteins. Two DNA vaccine constructs (IC1-90 and IC48) that differed by point mutations in gag and pol were compared. IC1-90 produces primarily immature (core comprises unprocessed Pr55Gag) HIV-like particles (VLPs) and IC48 produces mature VLP with processed Pr55Gag, immature VLP, and intracellular protein aggregates. Both vaccines raised significant cellular responses for Gag, Pol, and Env. Approximate twofold higher ELISPOT responses to Gag and Env epitopes were observed for IC48 animals than for IC1-90 animals at the peak post-MVA effector (P = 0.028) and late memory (P = 0.051) phases, respectively. Greater breadth for IC48-primed animals was observed than for IC1-90-primed animals at peak response (P = 0.03). Our results indicated that the vaccines elicited high frequency T cell responses and primed anti-Env antibody. They also suggest that expression of different forms of VLP has a significant effect on elicited cellular and humoral immunity.
Collapse
Affiliation(s)
- Dennis Ellenberger
- Laboratory Branch, Centers for Disease Control and Prevention, Mail Stop G-19, Atlanta, GA 30333, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Vaccination, or the deliberate induction of protective immunity by administering nonpathogenic forms of a microbe or its antigens to induce a memory immune response, is the world's most cost-effective medical procedure for preventing morbidity and mortality caused by infectious disease. Historically, most vaccines have worked by eliciting long-lived plasma cells. These cells produce antibodies that limit disease by neutralizing a toxin or blocking the spread of the infectious agent. For these 'B cell vaccines,' the immunological marker, or correlate, for protection is the titer of protective antibodies. With the discovery of HIV/AIDS, vaccine development has been confronted by an agent that is not easily blocked by antibody. To overcome this, researchers who are developing HIV/AIDS vaccines have turned to the elicitation of cellular immunity, or 'T cell vaccines,' which recognize and kill infected cells.
Collapse
Affiliation(s)
- Harriet L Robinson
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329, USA.
| | | |
Collapse
|
50
|
Amara RR, Sharma S, Patel M, Smith JM, Chennareddi L, Herndon JG, Robinson HL. Studies on the cross-clade and cross-species conservation of HIV-1 Gag-specific CD8 and CD4 T cell responses elicited by a clade B DNA/MVA vaccine in macaques. Virology 2005; 334:124-33. [PMID: 15749128 DOI: 10.1016/j.virol.2005.01.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2004] [Revised: 12/30/2004] [Accepted: 01/21/2005] [Indexed: 11/20/2022]
Abstract
Here, we evaluate the T cell responses raised by our HIV-1 clade B DNA/MVA vaccine for recognition of a HIV-1 circulating recombinant form (CRF) AG Gag sequence (CRF-02). The cross-clade activity for the AG sequence was better conserved for CD8 than CD4 T cells. CD8 T cells exhibited 75% conservation for height and 83% conservation for breadth, whereas CD4 responses exhibited 45% conservation for height and 50% conservation for breadth. Five CD8 epitopes and 8 CD4 epitopes were mapped. Three of the 5 CD8 epitopes and 2 of the 8 CD4 epitopes were conserved across multiple HIV-1 clades. Impressively, all of the CD8 epitopes and half of the CD4 epitopes have been reported for human infections. Our results demonstrate that the clade B DNA/MVA HIV vaccine elicits T cell responses against epitopes that are conserved in multiple clades and recognized by humans and macaques.
Collapse
Affiliation(s)
- Rama Rao Amara
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | |
Collapse
|