1
|
Shabani SH, Kardani K, Milani A, Bolhassani A. In Silico and in Vivo Analysis of HIV-1 Rev Regulatory Protein for Evaluation of a Multiepitope-based Vaccine Candidate. Immunol Invest 2021; 51:1-28. [PMID: 33416004 DOI: 10.1080/08820139.2020.1867163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In silico-designed multiepitope conserved regions of human immunodeficiency virus 1 (HIV-1) proteins would be a beneficial strategy for antigen design which induces effective anti-HIV-1 T-cell responses. The conserved multiple HLA-DR-binding epitopes of Rev protein were identified using IEDB MHC-I prediction tools and SYFPEITHI webserver to screen potential T-cell epitopes. We analyzed toxicity, allergenicity, immunogenicity, hemolytic activity, cross-reactivity, cell-penetrating peptide (CPP) potency, and molecular docking of the candidate epitopes using several immune-informatics tools. Afterward, we designed a novel multiepitope construct based on non-toxic and non-allergenic Rev, Nef, Gp160 and P24-derived cytotoxic T cell (CTL) and T-helper cell (HTL) epitopes. Next, the designed construct (Nef-Rev-Gp160-P24) was subjected to three B-cell epitope prediction webservers, ProtParam and Protein-Sol to obtain the physicochemical features. Then, the recombinant multiepitope DNA and polypeptide constructs were complexed with different CPPs for nanoparticle formation and pass them via the cell membranes. Finally, the immunogenicity of multiepitope constructs in a variety of modalities was evaluated in mice. The results demonstrated that groups immunized with heterologous DNA+ MPG or HR9 CPP prime/rNef-Rev-Gp160-P24 polypeptide + LDP-NLS CPP boost regimens could significantly produce higher levels of IFN-γ and Granzyme B, and lower amounts of IL-10 than other groups. Moreover, higher levels of IgG2a and IgG2b were observed in all heterologous prime-boost regimens than homologous DNA or polypeptide regimens. Altogether, the present findings indicated that the Nef-Rev-Gp160-P24 polypeptide meets the criteria to be potentially useful as a multiepitope-based vaccine candidate against HIV-1 infection.
Collapse
Affiliation(s)
- Samaneh H Shabani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
2
|
Talayev V, Zaichenko I, Svetlova M, Matveichev A, Babaykina O, Voronina E, Mironov A. Low-dose influenza vaccine Grippol Quadrivalent with adjuvant Polyoxidonium induces a T helper-2 mediated humoral immune response and increases NK cell activity. Vaccine 2020; 38:6645-6655. [PMID: 32873403 DOI: 10.1016/j.vaccine.2020.07.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/21/2020] [Accepted: 07/26/2020] [Indexed: 12/20/2022]
Abstract
The influenza vaccine Grippol® Quadrivalent (GQ) is a new vaccine, containing the adjuvant Polyoxidonium® and recombinant hemagglutinins from 4 strains of the influenza virus in amount of 5-6 μg of each hemagglutinin per human dose. These doses of antigens are about 3 times less than the standard dose recommended by WHO. We sought to characterize the immune response to the GQ vaccine and to determine the contribution of the adjuvant in this response. BALB/c mice were vaccinated with GQ or with adjuvant-free antigen mixtures (AGs). Then, the antibody response, the number of memory T cells in the spleen, and the functional properties of splenocytes were determined. The vaccine GQ has been shown to induce antibodies to all 4 influenza hemagglutinins. The vaccination with GQ caused a strong increase in the AG-induced proliferation and production of Th2 cytokines ex vivo. These effects were equal to effect achieved by standard dose of antigens. Vaccination also caused the accumulation of CD4+ large lymphocytes with the phenotype of central and effector memory T cells in the spleen. The GQ vaccine enhanced the cytolytic activity of natural killer (NK) cells, whereas the adjuvant-free mixture of AGs in lowered and standard doses did not affect NK activity. We did not find a noticeable response of Th1 and CD8+ T cells to vaccination. In vitro, the GQ vaccine stimulated the maturation of human monocyte-derived dendritic cells (DCs) enhancing the expression of HLA-DR, CD80, CD83, CD86 and ICOSL molecules. Polyoxidonium without AGs also induced expression of ICOSL, which plays an important role in T-dependent humoral immune response. In summary, the low-dose influenza vaccine GQ with Polyoxidonium adjuvant is immunogenic, induces a Th2-polarized T-cell response and CD4+ memory T cells maturation, activates the production of antibodies to influenza hemagglutinins, and increases the activity of NK cells.
Collapse
Affiliation(s)
- Vladimir Talayev
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 603950, 71 M. Yamskay str., Nizhny Novgorod, Russia.
| | - Irina Zaichenko
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 603950, 71 M. Yamskay str., Nizhny Novgorod, Russia
| | - Maria Svetlova
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 603950, 71 M. Yamskay str., Nizhny Novgorod, Russia
| | - Alexei Matveichev
- NPO Petrovax Pharm LLC, 142143, 1 Sosnovaya St., Pokrov Village, Podolsk, Moscow Region, Russia
| | - Olga Babaykina
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 603950, 71 M. Yamskay str., Nizhny Novgorod, Russia
| | - Elena Voronina
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 603950, 71 M. Yamskay str., Nizhny Novgorod, Russia
| | - Andrei Mironov
- Lobachevsky State University of Nizhny Novgorod, 603950, 23 Prospekt Gagarina (Gagarin Avenue), Nizhny Novgorod, Russia
| |
Collapse
|
3
|
Sarkar S, Piepenbrink MS, Basu M, Thakar J, Keefer MC, Hessell AJ, Haigwood NL, Kobie JJ. IL-33 enhances the kinetics and quality of the antibody response to a DNA and protein-based HIV-1 Env vaccine. Vaccine 2019; 37:2322-2330. [PMID: 30926296 PMCID: PMC6506229 DOI: 10.1016/j.vaccine.2019.03.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/05/2019] [Accepted: 03/21/2019] [Indexed: 12/12/2022]
Abstract
Induction of a sustained and broad antibody (Ab) response is a major goal in developing a protective HIV-1 vaccine. DNA priming alone shows reduced levels of immunogenicity; however, when combined with protein boosting is an attractive vaccination strategy for induction of humoral responses. Using the VC10014 DNA and protein-based vaccine consisting of HIV-1 envelope (Env) gp160 plasmids and trimeric gp140 proteins derived from an HIV-1 clade B infected subject who developed broadly neutralizing serum Abs, and which has been previously demonstrated to induce Tier 2 heterologous neutralizing Abs in rhesus macaques, we evaluated whether MPLA and IL-33 when administered during the DNA priming phase enhances the humoral response in mice. The addition of IL-33 during the gp160 DNA priming phase resulted in high titer gp120-specific plasma IgG after the first immunization. The IL-33 treated mice had higher plasma IgG Ab avidity, breadth, and durability after DNA and protein co-immunization with alum adjuvant as compared to MPLA and alum only treated mice. IL-33 was also associated with a significant IgM Env-specific response and expansion of peritoneal and splenic B-1b B cells. These results indicate that DNA priming in the presence of exogenous IL-33 qualitatively alters the HIV-1 Env-specific humoral response, improving the kinetics and breadth of potentially protective Ab.
Collapse
Affiliation(s)
- Sanghita Sarkar
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, NY, United States
| | - Michael S Piepenbrink
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, NY, United States
| | - Madhubanti Basu
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, NY, United States
| | - Juilee Thakar
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Michael C Keefer
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, NY, United States
| | - Ann J Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Nancy L Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - James J Kobie
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, NY, United States.
| |
Collapse
|
4
|
Fleck JD, Betti AH, da Silva FP, Troian EA, Olivaro C, Ferreira F, Verza SG. Saponins from Quillaja saponaria and Quillaja brasiliensis: Particular Chemical Characteristics and Biological Activities. Molecules 2019; 24:E171. [PMID: 30621160 PMCID: PMC6337100 DOI: 10.3390/molecules24010171] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/13/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022] Open
Abstract
Quillaja saponaria Molina represents the main source of saponins for industrial applications. Q. saponaria triterpenoids have been studied for more than four decades and their relevance is due to their biological activities, especially as a vaccine adjuvant and immunostimulant, which have led to important research in the field of vaccine development. These saponins, alone or incorporated into immunostimulating complexes (ISCOMs), are able to modulate immunity by increasing antigen uptake, stimulating cytotoxic T lymphocyte production (Th1) and cytokines (Th2) in response to different antigens. Furthermore, antiviral, antifungal, antibacterial, antiparasitic, and antitumor activities are also reported as important biological properties of Quillaja triterpenoids. Recently, other saponins from Q. brasiliensis (A. St.-Hill. & Tul.) Mart. were successfully tested and showed similar chemical and biological properties to those of Q. saponaria barks. The aim of this manuscript is to summarize the current advances in phytochemical and pharmacological knowledge of saponins from Quillaja plants, including the particular chemical characteristics of these triterpenoids. The potential applications of Quillaja saponins to stimulate further drug discovery research will be provided.
Collapse
Affiliation(s)
- Juliane Deise Fleck
- Molecular Microbiology Laboratory, Institute of Health Sciences, Feevale University, Novo Hamburgo 93525-075, RS, Brazil.
| | - Andresa Heemann Betti
- Bioanalysis Laboratory, Institute of Health Sciences, Feevale University, Novo Hamburgo 93525-075, RS, Brazil.
| | - Francini Pereira da Silva
- Molecular Microbiology Laboratory, Institute of Health Sciences, Feevale University, Novo Hamburgo 93525-075, RS, Brazil.
| | - Eduardo Artur Troian
- Molecular Microbiology Laboratory, Institute of Health Sciences, Feevale University, Novo Hamburgo 93525-075, RS, Brazil.
| | - Cristina Olivaro
- Science and Chemical Technology Department, University Center of Tacuarembó, Udelar, Tacuarembó 45000, Uruguay.
| | - Fernando Ferreira
- Organic Chemistry Department, Carbohydrates and Glycoconjugates Laboratory, Udelar, Mondevideo 11600, Uruguay.
| | - Simone Gasparin Verza
- Molecular Microbiology Laboratory, Institute of Health Sciences, Feevale University, Novo Hamburgo 93525-075, RS, Brazil.
| |
Collapse
|
5
|
Menon V, Ayala VI, Rangaswamy SP, Kalisz I, Whitney S, Galmin L, Ashraf A, LaBranche C, Montefiori D, Petrovsky N, Kalyanaraman VS, Pal R. DNA prime/protein boost vaccination elicits robust humoral response in rhesus macaques using oligomeric simian immunodeficiency virus envelope and Advax delta inulin adjuvant. J Gen Virol 2017; 98:2143-2155. [PMID: 28758637 DOI: 10.1099/jgv.0.000863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The partial success of the RV144 trial underscores the importance of envelope-specific antibody responses for an effective HIV-1 vaccine. Oligomeric HIV-1 envelope proteins delivered with a potent adjuvant are expected to elicit strong antibody responses with broad neutralization specificity. To test this hypothesis, two SIV envelope proteins were formulated with delta inulin-based adjuvant (Advax) and used to immunize nonhuman primates. Oligomeric gp140-gp145 from SIVmac251 and SIVsmE660 was purified to homogeneity. Oligomers showed high-affinity interaction with CD4 and were highly immunogenic in rabbits, inducing Tier 2 SIV-neutralizing antibodies. The immunogenicity of an oligomeric Env DNA prime and protein boost together with Advax was evaluated in Chinese rhesus macaques. DNA administration elicited antibodies to both envelopes, and titres were markedly enhanced following homologous protein boosts via intranasal and intramuscular routes. Strong antibody responses were detected against the V1 and V2 domains of gp120. During peak immune responses, a low to moderate level of neutralizing activity was detected against Tier 1A/1B SIV isolates, with a moderate level noted against a Tier 2 isolate. Increased serum antibody affinity to SIVmac251 gp140 and generation of Env-specific memory B cells were observed in the immunized macaques. Animals were subjected to low-dose intravaginal challenge with SIVmac251 one week after the last protein boost. One out of three immunized animals was protected from infection. Although performed with a small number of macaques, this study demonstrates the utility of oligomeric envelopes formulated with Advax in eliciting broad antibody responses with the potential to provide protection against SIV transmission.
Collapse
Affiliation(s)
- Veena Menon
- Advanced BioScience Laboratories, Inc., Rockville, MD, USA
| | - Victor I Ayala
- Advanced BioScience Laboratories, Inc., Rockville, MD, USA
| | | | - Irene Kalisz
- Advanced BioScience Laboratories, Inc., Rockville, MD, USA
| | | | - Lindsey Galmin
- Advanced BioScience Laboratories, Inc., Rockville, MD, USA
| | - Asma Ashraf
- Advanced BioScience Laboratories, Inc., Rockville, MD, USA
| | | | | | - Nikolai Petrovsky
- Vaxine Pty Ltd and Flinders University, Bedford Park, SA 5042, Australia
| | | | - Ranajit Pal
- Advanced BioScience Laboratories, Inc., Rockville, MD, USA
| |
Collapse
|
6
|
Pouliot K, Buglione-Corbett R, Marty-Roix R, Montminy-Paquette S, West K, Wang S, Lu S, Lien E. Contribution of TLR4 and MyD88 for adjuvant monophosphoryl lipid A (MPLA) activity in a DNA prime-protein boost HIV-1 vaccine. Vaccine 2014; 32:5049-56. [PMID: 25045815 PMCID: PMC10687719 DOI: 10.1016/j.vaccine.2014.07.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 06/17/2014] [Accepted: 07/08/2014] [Indexed: 12/19/2022]
Abstract
Recombinant protein vaccines are commonly formulated with an immune-stimulatory compound, or adjuvant, to boost immune responses to a particular antigen. Recent studies have shown that, through recognition of molecular motifs, receptors of the innate immune system are involved in the functions of adjuvants to generate and direct adaptive immune responses. However, it is not clear to which degree those receptors are also important when the adjuvant is used as part of a novel heterologous prime-boost immunization process in which the priming and boosting components are not the same type of vaccines. In the current study, we compared the immune responses elicited by a pentavalent HIV-1 DNA prime-protein boost vaccine in mice deficient in either Toll-like receptor 4 (TLR4) or myeloid differentiation primary response gene 88 (MyD88) to wildtype mice. HIV gp120 protein administered in the boost phase was formulated with either monophosphoryl lipid A (MPLA), QS-21, or Al(OH)3. Endpoint antibody titer, serum cytokine response and T-cell memory response were assessed. Neither TLR4 nor MyD88 deficiency had a significant effect on the immune response of mice given vaccine formulated with QS-21 or Al(OH)3. However, TLR4- and MyD88-deficiency decreased both the antibody and T-cell responses in mice administered HIV gp120 formulated with MPLA. These results further our understanding of the activation of TLR4 and MyD88 by MPLA in the context of a DNA prime/protein boost immunization strategy.
Collapse
Affiliation(s)
- Kimberly Pouliot
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Rachel Buglione-Corbett
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Robyn Marty-Roix
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Sara Montminy-Paquette
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Kim West
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Egil Lien
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States; Centre of Molecular Inflammation Research, Dept. of Cancer and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| |
Collapse
|
7
|
Lewis B, Whitney S, Hudacik L, Galmin L, Huaman MC, Cristillo AD. Nedd4-mediated increase in HIV-1 Gag and Env proteins and immunity following DNA-vaccination of BALB/c mice. PLoS One 2014; 9:e91267. [PMID: 24614057 PMCID: PMC3948788 DOI: 10.1371/journal.pone.0091267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/10/2014] [Indexed: 02/02/2023] Open
Abstract
The late assembly domain of many viruses is critical for budding. Within these domains, encoded in viral structural proteins, are the conserved motifs PTAP, PPxY and YPxL. These sequences are the key determinants for association of viral proteins with intracellular molecules such as Tsg101, Nedd4 and AIP1/ALIX. While roles for Tsg101 and AIP1/ALIX in HIV-1 budding have been well established, less is known about the role of Nedd4. Recent studies, however, have identified a function for Nedd4-like protein in HIV-1 release. In this study, we investigated post-transcriptional changes of Nedd4 following SHIVSF162P3 infection of rhesus macaques, its role on HIV-1 p24 and gp120 levels in vitro and its potential as an immune modulator in HIV vaccination of BALB/c mice. Increased Nedd4 protein levels were noted in both CD4+ and CD8+ T cells following SHIVSF162P3-infection of naïve macaques. Transient co-transfection studies in 293 cells with HXB2 and Nedd4 demonstrated a Nedd4-mediated increase in p24 and gp120 levels. This increase was found to be dependent on the Ca2+/calmodulin-regulated phospholipid binding C2 domain and not ubiquitin ligase activity or HIV LTR activity. Co-transfection of Nedd4 with plasmid DNA expressing Gag or Env was further shown to augment both intracellular and extracellular Gag or Env proteins. To assess the potential of Nedd4 as an immune modulator, BALB/c mice were immunized intramuscularly with plasmid DNA encoding HIV gag, env and Nedd4. Nedd4 co-administration was found to increase serum anti-p24 but not anti-gp120 antibodies. Nedd4 co-injection was found to have no affect on Gag- or Env-specific IFNγ but had a trend of increased Gag-specific IL-6, IL-17A and TNFα that was not seen following Env stimulation. Based on our initial findings, Nedd4-mediated changes in HIV protein levels and its potential use in HIV-1 vaccine development warrants further investigation.
Collapse
Affiliation(s)
- Brad Lewis
- Advanced BioScience Laboratories, Inc., Rockville, Maryland, United States of America
| | - Stephen Whitney
- Advanced BioScience Laboratories, Inc., Rockville, Maryland, United States of America
| | - Lauren Hudacik
- Advanced BioScience Laboratories, Inc., Rockville, Maryland, United States of America
| | - Lindsey Galmin
- Advanced BioScience Laboratories, Inc., Rockville, Maryland, United States of America
| | - Maria Cecilia Huaman
- Advanced BioScience Laboratories, Inc., Rockville, Maryland, United States of America
| | - Anthony D. Cristillo
- Advanced BioScience Laboratories, Inc., Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
8
|
Abstract
Vaccines have evolved for hundreds of years, but all utilize the premise that safely pre-exposing the host to some component of a pathogen allows for enhanced immune recognition, and potential protection from disease, upon encountering the pathogen at a later date. Early vaccination strategies used inactivated or attenuated vaccines, many of which contained toxins and other components that resulted in reactogenicity or risk of reversion to virulence. DNA vaccines supplant many of the issues associated with inactivated or attenuated vaccines, but these vaccines tend to provide weak immunological responses, particularly in primates. DNA Electroporation may prove to be the "missing link" in the evolution of DNA vaccines allowing for enhanced immune responses from DNA vaccination in humans thereby resulting in protection from disease post-pathogen exposure.
Collapse
|
9
|
Li W, Wang S, Lu S. Pilot Study on the Use of DNA Priming Immunization to Enhance Y. pestis LcrV-Specific B Cell Responses Elicited by a Recombinant LcrV Protein Vaccine. Vaccines (Basel) 2013; 2:36-48. [PMID: 26344467 PMCID: PMC4494201 DOI: 10.3390/vaccines2010036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/26/2013] [Accepted: 12/05/2013] [Indexed: 11/17/2022] Open
Abstract
Recent studies indicate that DNA immunization is powerful in eliciting antigen-specific antibody responses in both animal and human studies. However, there is limited information on the mechanism of this effect. In particular, it is not known whether DNA immunization can also enhance the development of antigen-specific B cell development. In this report, a pilot study was conducted using plague LcrV immunogen as a model system to determine whether DNA immunization is able to enhance LcrV-specific B cell development in mice. Plague is an acute and often fatal infectious disease caused by Yersinia pestis (Y. pestis). Humoral immune responses provide critical protective immunity against plague. Previously, we demonstrated that a DNA vaccine expressing LcrV antigen can protect mice from lethal mucosal challenge. In the current study, we further evaluated whether the use of a DNA priming immunization is able to enhance the immunogenicity of a recombinant LcrV protein vaccine, and in particular, the development of LcrV-specific B cells. Our data indicate that DNA immunization was able to elicit high-level LcrV antibody responses when used alone or as part of a prime-boost immunization approach. Most significantly, DNA immunization was also able to increase the levels of LcrV-specific B cell development. The finding that DNA immunization can enhance antigen-specific B cell responses is highly significant and will help guide similar studies in other model antigen systems.
Collapse
Affiliation(s)
- Wei Li
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Shixia Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Shan Lu
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
10
|
Abstract
DNA immunization was discovered in early 1990s, and its use has been expanded from vaccine studies to a broader range of biomedical research areas, such as the generation of high-quality polyclonal and monoclonal antibodies as research reagents. In this unit, three common DNA immunization methods are described: needle injection, electroporation, and gene gun. In addition, several common considerations related to DNA immunization are discussed.
Collapse
Affiliation(s)
- Shixia Wang
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Shan Lu
- University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
11
|
Buglione-Corbett R, Pouliot K, Marty-Roix R, West K, Wang S, Lien E, Lu S. Serum cytokine profiles associated with specific adjuvants used in a DNA prime-protein boost vaccination strategy. PLoS One 2013; 8:e74820. [PMID: 24019983 PMCID: PMC3760864 DOI: 10.1371/journal.pone.0074820] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/05/2013] [Indexed: 11/30/2022] Open
Abstract
In recent years, heterologous prime-boost vaccines have been demonstrated to be an effective strategy for generating protective immunity, consisting of both humoral and cell-mediated immune responses against a variety of pathogens including HIV-1. Previous reports of preclinical and clinical studies have shown the enhanced immunogenicity of viral vector or DNA vaccination followed by heterologous protein boost, compared to using either prime or boost components alone. With such approaches, the selection of an adjuvant for inclusion in the protein boost component is expected to impact the immunogenicity and safety of a vaccine. In this study, we examined in a mouse model the serum cytokine and chemokine profiles for several candidate adjuvants: QS-21, Al(OH)3, monophosphoryl lipid A (MPLA) and ISCOMATRIX™ adjuvant, in the context of a previously tested pentavalent HIV-1 Env DNA prime-protein boost formulation, DP6-001. Our data revealed that the candidate adjuvants in the context of the DP6-001 formulation are characterized by unique serum cytokine and chemokine profiles. Such information will provide valuable guidance in the selection of an adjuvant for future AIDS vaccine development, with the ultimate goal of enhancing immunogenicity while minimizing reactogenicity associated with the use of an adjuvant. More significantly, results reported here will add to the knowledge on how to include an adjuvant in the context of a heterologous prime-protein boost vaccination strategy in general.
Collapse
Affiliation(s)
- Rachel Buglione-Corbett
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kimberly Pouliot
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Robyn Marty-Roix
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kim West
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Shixia Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Egil Lien
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Shan Lu
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
12
|
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
|
13
|
Hallengärd D, Applequist SE, Nyström S, Maltais AK, Marovich M, Moss B, Earl P, Nihlmark K, Wahren B, Bråve A. Immunization with multiple vaccine modalities induce strong HIV-specific cellular and humoral immune responses. Viral Immunol 2012; 25:423-32. [PMID: 23035853 PMCID: PMC10970668 DOI: 10.1089/vim.2012.0046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/05/2012] [Indexed: 12/30/2022] Open
Abstract
Heterologous priming and boosting with antigens expressed by DNA, viral vectors, or as proteins, are experimental strategies to induce strong immune responses against infectious diseases and cancer. In a preclinical study we compared the ability of recombinant modified vaccinia Ankara encoding HIV antigens (MVA-CMDR), and/or recombinant gp140C (rgp140C), to boost responses induced by a multigene/multisubtype HIV DNA vaccine delivered by electroporation (EP). Homologous DNA immunizations augmented by EP stimulated strong cellular immune responses. Still stronger cellular immune responses were observed after DNA priming and MVA-CMDR boosting, which was superior to all other immunization schedules tested in terms of antigen-specific IFN-γ, IL-2, and bifunctional IFN-γ and IL-2 responses. For HIV Env-specific antibody responses, mice receiving repeated rgp140C immunizations, and mice boosted with rgp140C, elicited the highest binding titers and the highest numbers of antibody-secreting B cells. When considering both cellular and humoral immune responses, a combination of DNA, MVA-CMDR, and rgp140C immunizations induced the overall most potent immune responses and the highest avidity of HIV Env-specific antibodies. These data emphasize the importance of including multiple vaccine modalities that can stimulate both T and B cells, and thus elicit strong and balanced immune responses. The present HIV vaccine combination holds promise for further evaluation in clinical trials.
Collapse
Affiliation(s)
- David Hallengärd
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Yuan T, Li J, Zhang Y, Wang Y, Streaker E, Dimitrov DS, Zhang MY. Putative rhesus macaque germline predecessors of human broadly HIV-neutralizing antibodies: differences from the human counterparts and implications for HIV-1 vaccine development. Vaccine 2011; 29:6903-10. [PMID: 21807049 DOI: 10.1016/j.vaccine.2011.07.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/08/2011] [Accepted: 07/13/2011] [Indexed: 10/17/2022]
Abstract
Broadly neutralizing antibodies (bnAbs) are likely to be a key component of protective immunity conferred by an effective HIV-1 vaccine. We and others have reported that putative human germline predecessors of known human bnAbs lack measurable binding to HIV-1 envelope glycoproteins (Env), which could be a new challenge for eliciting human bnAbs. Rhesus macaques have been used as nonhuman primate models for testing vaccine candidates, but little is known about their germline Abs. Here we show the similarities and differences between putative rhesus macaque and human germline predecessors and possible intermediate antibodies of one of the best characterized bnAbs, b12. Similar to the human counterpart, a putative rhesus macaque b12 germline antibody lacks measurable binding to HIV-1 Envs, suggesting that initiation of somatic maturation of rhesus macaque germline b12 predecessor may also be a challenge. However, differences in sequence characteristics and binding properties between macaque and human b12 germline and intermediate antibodies suggest that the two germline predecessors may undergo different maturation pathways in rhesus macaques and in humans. These results indicate that immunogens that could initiate the immune responses and drive somatic mutations leading to elicitation of b12 or b12-like bnAbs in rhesus macaques and in humans are likely to be different. This has important implications for HIV-1 vaccine development.
Collapse
Affiliation(s)
- Tingting Yuan
- AIDS Institute, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Cristillo AD, Ferrari MG, Hudacik L, Lewis B, Galmin L, Bowen B, Thompson D, Petrovsky N, Markham P, Pal R. Induction of mucosal and systemic antibody and T-cell responses following prime-boost immunization with novel adjuvanted human immunodeficiency virus-1-vaccine formulations. J Gen Virol 2011; 92:128-40. [PMID: 21169215 PMCID: PMC3052530 DOI: 10.1099/vir.0.023242-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
As sexual transmission of human immunodeficiency virus-1 (HIV-1) occurs via the mucosa, an ideal HIV-1 vaccine should induce both mucosal and systemic immunity. We therefore sought to evaluate the induction of mucosal responses using a DNA env prime–gp120 protein boost approach in which sequential nasal and parenteral protein administration was performed with two novel carbohydrate-based adjuvants. These adjuvants, Advax-M and Advax-P, were specifically designed for mucosal and systemic immune enhancement, respectively. Murine intranasal immunization with gp120/Advax-M adjuvant elicited gp120-specific IgA in serum and mucosal secretions that was markedly enhanced by DNA priming. Boosting of DNA-primed mice with gp120/Advax-M and gp120/Advax-P by sequential intranasal and intramuscular immunization, or vice versa, elicited persistent mucosal gp120-specific IgA, systemic IgG and memory T- and B-cell responses. Induction of homologous, but not heterologous, neutralizing activity was noted in the sera of all immunized groups. While confirmation of efficacy is required in challenge studies using non-human primates, these results suggest that the combination of DNA priming with sequential nasal and parenteral protein boosting, with appropriate mucosal and systemic adjuvants, could generate strong mucosal and systemic immunity and may block HIV-1 mucosal transmission and infection.
Collapse
Affiliation(s)
- Anthony D Cristillo
- Advanced BioScience Laboratories Inc., 5510 Nicholson Lane, Kensington, MD 20895, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
A major hurdle in the development of a global HIV-1 vaccine is viral diversity. For close to three decades, HIV vaccine development has focused on either the induction of T cell immune responses or antibody responses, and only rarely on both components. After the failure of the STEP trial, the scientific community concluded that a T cell-based vaccine would likely not be protective if the T cell immune responses were elicited against only a few dominant epitopes. Similarly, for vaccines focusing on antibody responses, one of the main criticisms after VaxGen's failed Phase III trials was on the limited antigen breadth included in the two formulations used. The successes of polyvalent vaccine approaches against other antigenically variable pathogens encourage implementation of the same approach for the design of HIV-1 vaccines. A review of the existing HIV-1 vaccination approaches based on the polyvalent principle is included here to provide a historical perspective for the current effort of developing a polyvalent HIV-1 vaccine. Results summarized in this review provide a clear indication that the polyvalent approach is a viable one for the future development of an effective HIV vaccine.
Collapse
Affiliation(s)
- Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | | | |
Collapse
|
17
|
Innovative bioinformatic approaches for developing peptide-based vaccines against hypervariable viruses. Immunol Cell Biol 2010; 89:81-9. [PMID: 20458336 DOI: 10.1038/icb.2010.65] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The application of the fields of pharmacogenomics and pharmacogenetics to vaccine design has been recently labeled 'vaccinomics'. This newly named area of vaccine research, heavily intertwined with bioinformatics, seems to be leading the charge in developing novel vaccines for currently unmet medical needs against hypervariable viruses such as human immunodeficiency virus (HIV), hepatitis C and emerging avian and swine influenza. Some of the more recent bioinformatic approaches in the area of vaccine research include the use of epitope determination and prediction algorithms for exploring the use of peptide epitopes as vaccine immunogens. This paper briefly discusses and explores some current uses of bioinformatics in vaccine design toward the pursuit of peptide vaccines for hypervariable viruses. The various informatics and vaccine design strategies attempted by other groups toward hypervariable viruses will also be briefly examined, along with the strategy used by our group in the design and synthesis of peptide immunogens for candidate HIV and influenza vaccines.
Collapse
|
18
|
Zhao HP, Sun JF, Li N, Sun Y, Wang Y, Qiu HJ. Prime-boost immunization using alphavirus replicon and adenovirus vectored vaccines induces enhanced immune responses against classical swine fever virus in mice. Vet Immunol Immunopathol 2009; 131:158-66. [DOI: 10.1016/j.vetimm.2009.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 03/30/2009] [Accepted: 04/03/2009] [Indexed: 02/06/2023]
|
19
|
CT043, a protective antigen that induces a CD4+ Th1 response during Chlamydia trachomatis infection in mice and humans. Infect Immun 2009; 77:4168-76. [PMID: 19596772 DOI: 10.1128/iai.00344-09] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Despite several decades of intensive studies, no vaccines against Chlamydia trachomatis, an intracellular pathogen causing serious ocular and urogenital diseases, are available yet. Infection-induced immunity in both animal models and humans strongly supports the notion that for a vaccine to be effective a strong CD4(+) Th1 immune response should be induced. In the course of our vaccine screening program based on the selection of chlamydial proteins eliciting cell-mediated immunity, we have found that CT043, a protein annotated as hypothetical, induces CD4(+) Th1 cells both in chlamydia-infected mice and in human patients with diagnosed C. trachomatis genital infection. DNA priming/protein boost immunization with CT043 results in a 2.6-log inclusion-forming unit reduction in the murine lung infection model. Sequence analysis of CT043 from C. trachomatis human isolates belonging to the most representative genital serovars revealed a high degree of conservation, suggesting that this antigen could provide cross-serotype protection. Therefore, CT043 is a promising vaccine candidate against C. trachomatis infection.
Collapse
|
20
|
Bodles-Brakhop AM, Heller R, Draghia-Akli R. Electroporation for the delivery of DNA-based vaccines and immunotherapeutics: current clinical developments. Mol Ther 2009; 17:585-92. [PMID: 19223870 PMCID: PMC2835112 DOI: 10.1038/mt.2009.5] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 12/27/2008] [Indexed: 11/09/2022] Open
Abstract
Electroporation (EP) has been used in basic research for the past 25 years to aid in the transfer of DNA into cells in vitro. EP in vivo enhances transfer of DNA vaccines and therapeutic plasmids to the skin, muscle, tumors, and other tissues resulting in high levels of expression, often with serological and clinical benefits. The recent interest in nonviral gene transfer as treatment options for a vast array of conditions has resulted in the refinement and optimization of EP technology. Current research has revealed that EP can be successfully used in many species, including humans. Clinical trials are currently under way. Herein, the transition of EP from basic science to clinical trials will be discussed.
Collapse
Affiliation(s)
- Angela M Bodles-Brakhop
- VGX Pharmaceuticals, Inc., 2700 Research Forest Drive, Suite 180, The Woodlands, Texas 77381, USA.
| | | | | |
Collapse
|
21
|
McBurney SP, Ross TM. Viral sequence diversity: challenges for AIDS vaccine designs. Expert Rev Vaccines 2008; 7:1405-17. [PMID: 18980542 DOI: 10.1586/14760584.7.9.1405] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Among the greatest challenges facing AIDS vaccine development is the intrinsic diversity among circulating populations of HIV-1 in various geographical locations and the need to develop vaccines that can elicit enduring protective immunity to variant HIV-1 strains. While variation is observed in all of the viral proteins, the greatest diversity is localized to the viral envelope glycoproteins, evidently reflecting the predominant role of these proteins in eliciting host immune recognition and responses that result in progressive evolution of the envelope proteins during persistent infection. Interestingly, while envelope glycoprotein variation is widely assumed to be a major obstacle to AIDS vaccine development, there is very little experimental data in animal or human lentivirus systems addressing this critical issue. In this review, the state of vaccine development to address envelope diversity will be presented, focusing on the use of centralized and polyvalent sequence design as mechanisms to elicit broadly reactive immune responses.
Collapse
Affiliation(s)
- Sean P McBurney
- University of Pittsburgh, School of Medicine, Center for Vaccine Research, Program in Molecular Virology and Microbiology, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
22
|
Patterson LJ, Robert-Guroff M. Replicating adenovirus vector prime/protein boost strategies for HIV vaccine development. Expert Opin Biol Ther 2008; 8:1347-63. [PMID: 18694354 DOI: 10.1517/14712598.8.9.1347] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND In recent years the HIV vaccine field introduced a number of promising vaccine candidates into human clinical trials. OBJECTIVE To briefly discuss the advances made in vaccine development and HIV pathogenesis and give an overview of the body of work our lab has generated in multiple animal models on replication-competent Adenovirus recombinant vaccines. METHODS Emphasis is placed on comparative examination of vaccine components, routes of immunization and challenge models using replicating Adenovirus vectors. RESULTS/CONCLUSION The findings make the case that replicating Adenovirus vectors are superior in priming multiple arms of the immune system, and in conjunction with protein boosting, have resulted in dramatic protective efficacy leading to their advancement to Phase I trials. Implications of the recent halting of the Merck Ad5-HIV Phase IIb clinical trial of our vaccine approach and other vectored vaccines are discussed.
Collapse
Affiliation(s)
- L Jean Patterson
- National Cancer Institute, National Institutes of Health, Vaccine Branch, Bethesda, Maryland 20892-5065, USA
| | | |
Collapse
|
23
|
Wang S, Kennedy JS, West K, Montefiori DC, Coley S, Lawrence J, Shen S, Green S, Rothman AL, Ennis FA, Arthos J, Pal R, Markham P, Lu S. Cross-subtype antibody and cellular immune responses induced by a polyvalent DNA prime-protein boost HIV-1 vaccine in healthy human volunteers. Vaccine 2008; 26:3947-57. [PMID: 18724414 PMCID: PMC3743087 DOI: 10.1016/j.vaccine.2007.12.060] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
An optimally effective AIDS vaccine would likely require the induction of both neutralizing antibody and cell-mediated immune responses, which has proven difficult to obtain in previous clinical trials. Here we report on the induction of human immunodeficiency virus type-1 (HIV-1)-specific immune responses in healthy adult volunteers that received the multi-gene, polyvalent, DNA prime-protein boost HIV-1 vaccine formulation, DP6-001, in a Phase I clinical trial. Robust cross-subtype HIV-1 specific T cell responses were detected in IFN-gamma ELISPOT assays. Furthermore, we detected high titer serum antibody responses that recognized a wide range of primary HIV-1 Env antigens and also neutralized pseudotyped viruses that express the primary Env antigens from multiple HIV-1 subtypes. These findings demonstrate that the DNA prime-protein boost approach is an effective immunization method to elicit both humoral and cell-mediated immune responses in humans, and that a polyvalent Env formulation could generate broad immune responses against HIV-1 viruses with diverse genetic backgrounds.
Collapse
Affiliation(s)
- Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - Jeffrey S. Kennedy
- Center for Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - Kim West
- Center for Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - David C. Montefiori
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, United States
| | - Scott Coley
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - John Lawrence
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - Siyuan Shen
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - Sharone Green
- Center for Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - Alan L. Rothman
- Center for Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - Francis A. Ennis
- Center for Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda 20892, United States
| | - Ranajit Pal
- Department of Cell Biology, Advanced BioScience Laboratories, Kensington, MD 20895, United States
| | - Phillip Markham
- Department of Cell Biology, Advanced BioScience Laboratories, Kensington, MD 20895, United States
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| |
Collapse
|
24
|
Letellier C, Boxus M, Rosar L, Toussaint JF, Walravens K, Roels S, Meyer G, Letesson JJ, Kerkhofs P. Vaccination of calves using the BRSV nucleocapsid protein in a DNA prime-protein boost strategy stimulates cell-mediated immunity and protects the lungs against BRSV replication and pathology. Vaccine 2008; 26:4840-8. [PMID: 18644416 PMCID: PMC7115630 DOI: 10.1016/j.vaccine.2008.06.100] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Revised: 06/25/2008] [Accepted: 06/29/2008] [Indexed: 11/06/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of respiratory disease in both cattle and young children. Despite the development of vaccines against bovine (B)RSV, incomplete protection and exacerbation of subsequent RSV disease have occurred. In order to circumvent these problems, calves were vaccinated with the nucleocapsid protein, known to be a major target of CD8+ T cells in cattle. This was performed according to a DNA prime–protein boost strategy. The results showed that DNA vaccination primed a specific T-cell-mediated response, as indicated by both a lymphoproliferative response and IFN-γ production. These responses were enhanced after protein boost. After challenge, mock-vaccinated calves displayed gross pneumonic lesions and viral replication in the lungs. In contrast, calves vaccinated by successive administrations of plasmid DNA and protein exhibited protection against the development of pneumonic lesions and the viral replication in the BAL fluids and the lungs. The protection correlated to the cell-mediated immunity and not to the antibody response.
Collapse
|
25
|
Multifunctional T-cell characteristics induced by a polyvalent DNA prime/protein boost human immunodeficiency virus type 1 vaccine regimen given to healthy adults are dependent on the route and dose of administration. J Virol 2008; 82:6458-69. [PMID: 18448544 DOI: 10.1128/jvi.00068-08] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A phase I clinical vaccine study of a human immunodeficiency virus type 1 (HIV-1) vaccine regimen comprising a DNA prime formulation (5-valent env and monovalent gag) followed by a 5-valent Env protein boost for seronegative adults was previously shown to induce HIV-1-specific T cells and anti-Env antibodies capable of neutralizing cross-clade viral isolates. In light of these initial findings, we sought to more fully characterize the HIV-1-specific T cells by using polychromatic flow cytometry. Three groups of participants were vaccinated three times with 1.2 mg of DNA administered intradermally (i.d.; group A), 1.2 mg of DNA administered intramuscularly (i.m.; group B), or 7.2 mg of DNA administered i.m. (high-dose group C) each time. Each group subsequently received one or two doses of 0.375 mg each of the gp120 protein boost vaccine (i.m.). Env-specific CD4 T-cell responses were seen in the majority of participants; however, the kinetics of responses differed depending on the route of DNA administration. The high i.m. dose induced the responses of the greatest magnitude after the DNA vaccinations, while the i.d. group exhibited the responses of the least magnitude. Nevertheless, after the second protein boost, the magnitude of CD4 T-cell responses in the i.d. group was indistinguishable from those in the other two groups. After the DNA vaccinations and the first protein boost, a greater number of polyfunctional Env-specific CD4 T cells (those with > or = 2 functions) were seen in the high-dose group than in the other groups. Gag-specific CD4 T cells and Env-specific CD8 T cells were seen only in the high-dose group. These findings demonstrate that the route and dose of DNA vaccines significantly impact the quality of immune responses, yielding important information for future vaccine design.
Collapse
|
26
|
Cristillo AD, Galmin L, Restrepo S, Hudacik L, Suschak J, Lewis B, Draghia-Akli R, Aziz N, Weiss D, Markham P, Pal R. HIV-1 Env vaccine comprised of electroporated DNA and protein co-administered with Talabostat. Biochem Biophys Res Commun 2008; 370:22-6. [PMID: 18329382 DOI: 10.1016/j.bbrc.2008.02.145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 02/29/2008] [Indexed: 11/26/2022]
Abstract
Selection of potent yet low reactogenic adjuvants for protein immunization is important for HIV-1 vaccine development. Immunogenicity of electroporated DNA (HIV env) and recombinant gp120, administered with either QS-21 or the orally administered immunomodulator, Talabostat, was evaluated in BALB/c mice. Electroporation of low dose DNA elicited Th1 cytokines and anti-envelope antibodies. Immunization with gp120 protein alone with or without Talabostat elicited lower Th1 and Th2 cytokine levels but comparable anti-gp120 antibodies to QS-21-formulated protein. Boosting of DNA-primed mice with gp120/Talabostat induced similar anti-gp120 antibody titers and slightly higher levels of Th1 and Th2 cytokines relative to QS-21-formulated protein. Induction of CD8(+) and CD4(+) T cells and functional CTL activity was noted. These results highlight the potential use of orally administered Talabostat for efficient protein boosting of antibody and T-cell responses primed by DNA.
Collapse
Affiliation(s)
- Anthony D Cristillo
- Advanced BioScience Laboratories, Inc., 5510 Nicholson Lane, Kensington, MD 20895, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang S, Kennedy JS, West K, Montefiori DC, Coley S, Lawrence J, Shen S, Green S, Rothman AL, Ennis FA, Arthos J, Pal R, Markham P, Lu S. Cross-subtype antibody and cellular immune responses induced by a polyvalent DNA prime-protein boost HIV-1 vaccine in healthy human volunteers. Vaccine 2008; 26:1098-110. [PMID: 18243434 PMCID: PMC2288749 DOI: 10.1016/j.vaccine.2007.12.024] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Revised: 12/03/2007] [Accepted: 12/13/2007] [Indexed: 01/01/2023]
Abstract
An optimally effective AIDS vaccine would likely require the induction of both neutralizing antibody and cell-mediated immune responses, which has proven difficult to obtain in previous clinical trials. Here we report on the induction of Human Immunodeficiency Virus Type-1 (HIV-1)-specific immune responses in healthy adult volunteers that received the multi-gene, polyvalent, DNA prime-protein boost HIV-1 vaccine formulation, DP6-001, in a Phase I clinical trial conducted in healthy adult volunteers of both genders. Robust cross-subtype HIV-1-specific T cell responses were detected in IFNgamma ELISPOT assays. Furthermore, we detected high titer serum antibody responses that recognized a wide range of primary HIV-1 Env antigens and also neutralized pseudotyped viruses that express the primary Env antigens from multiple HIV-1 subtypes. These findings demonstrate that the DNA prime-protein boost approach is an effective immunization method to elicit both humoral and cell-mediated immune responses in humans, and that a polyvalent Env formulation could generate broad immune responses against HIV-1 viruses with diverse genetic backgrounds.
Collapse
Affiliation(s)
- Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jeffrey S. Kennedy
- Center of Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Kim West
- Center of Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | | | - Scott Coley
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - John Lawrence
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Siyuan Shen
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Sharone Green
- Center of Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Alan L. Rothman
- Center of Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Francis A. Ennis
- Center of Infectious Disease and Vaccine Research, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, 20892
| | - Ranajit Pal
- Department of Cell Biology, Advanced BioScience Laboratories, Kensington, MD 20895
| | - Phillip Markham
- Department of Cell Biology, Advanced BioScience Laboratories, Kensington, MD 20895
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| |
Collapse
|
28
|
Patterson LJ, Beal J, Demberg T, Florese RH, Malkevich N, Venzon D, Aldrich K, Richardson E, Kalyanaraman VS, Kalisz I, Lee EM, Montefiori DC, Robey FA, Robert-Guroff M. Replicating adenovirus HIV/SIV recombinant priming alone or in combination with a gp140 protein boost results in significant control of viremia following a SHIV89.6P challenge in Mamu-A*01 negative rhesus macaques. Virology 2008; 374:322-37. [PMID: 18252262 DOI: 10.1016/j.virol.2007.12.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 11/13/2007] [Accepted: 12/24/2007] [Indexed: 11/27/2022]
Abstract
Previously, replicating adenovirus type 5 host range (Ad5hr)-HIV/SIV recombinant priming in combination with SIV envelope boosting, resulted in significant, durable protection in 39% of rhesus macaques after SIVmac251 challenge. Both Env-specific antibody mediating ADCC, and cellular immunity correlated with protection. Here we evaluate the relative immunogenicities of novel HIV proteins and their contribution to protection in a SHIV89.6P model. All groups were primed with Ad-HIVenv89.6P, SIVgag239, and SIVnef239 recombinants. One group was not boosted, one received HIV89.6Pgp140DeltaCFI protein, and one a novel HIV-1 poly-peptide "peptomer". The HIV89.6Pgp140DeltaCFI protein in adjuvant strongly boosted Env-specific antibody and memory T cell responses in blood and tissue, resulting in significant reductions in acute and set point viremia. Macaques not boosted, showed a significant reduction in set point viremia, a full 32 weeks after the last Ad priming immunization. The HIV peptomer-boosted group showed a trend toward chronic viremia reduction, but was not protected.
Collapse
Affiliation(s)
- L Jean Patterson
- Vaccine Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cristillo AD, Weiss D, Hudacik L, Restrepo S, Galmin L, Suschak J, Draghia-Akli R, Markham P, Pal R. Persistent antibody and T cell responses induced by HIV-1 DNA vaccine delivered by electroporation. Biochem Biophys Res Commun 2008; 366:29-35. [DOI: 10.1016/j.bbrc.2007.11.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 11/10/2007] [Indexed: 10/22/2022]
|
30
|
Koopman G, Mortier D, Hofman S, Mathy N, Koutsoukos M, Ertl P, Overend P, van Wely C, Thomsen LL, Wahren B, Voss G, Heeney JL. Immune-response profiles induced by human immunodeficiency virus type 1 vaccine DNA, protein or mixed-modality immunization: increased protection from pathogenic simian–human immunodeficiency virus viraemia with protein/DNA combination. J Gen Virol 2008; 89:540-5533. [PMID: 18198386 DOI: 10.1099/vir.0.83384-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Current data suggest that prophylactic human immunodeficiency virus type 1 (HIV) vaccines will be most efficacious if they elicit a combination of adaptive humoral and T-cell responses. Here, we explored the use of different vaccine strategies in heterologous prime–boost regimes and evaluated the breadth and nature of immune responses in rhesus monkeys induced by epidermally delivered plasmid DNA or recombinant HIV proteins formulated in the AS02A adjuvant system. These immunogens were administered alone or as either prime or boost in mixed-modality regimes. DNA immunization alone induced cell-mediated immune (CMI) responses, with a strong bias towards Th1-type cytokines, and no detectable antibodies to the vaccine antigens. Whenever adjuvanted protein was used as a vaccine, either alone or in a regime combined with DNA, high-titre antibody responses to all vaccine antigens were detected in addition to strong Th1- and Th2-type CMI responses. As the vaccine antigens included HIV-1 Env, Nef and Tat, as well as simian immunodeficiency virus (SIV)mac239 Nef, the animals were subsequently exposed to a heterologous, pathogenic simian–human immunodeficiency virus (SHIV)89.6p challenge. Protection against sustained high virus load was observed to some degree in all vaccinated groups. Suppression of virus replication to levels below detection was observed most frequently in the group immunized with protein followed by DNA immunization, and similarly in the group immunized with DNA alone. Interestingly, control of virus replication was associated with increased SIV Nef- and Gag-specific gamma interferon responses observed immediately following challenge.
Collapse
MESH Headings
- AIDS Vaccines/immunology
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/pharmacology
- Gene Products, env/genetics
- Gene Products, env/immunology
- Gene Products, env/metabolism
- Gene Products, tat/genetics
- Gene Products, tat/immunology
- Gene Products, tat/metabolism
- HIV/genetics
- HIV/immunology
- HIV/metabolism
- Human Immunodeficiency Virus Proteins/administration & dosage
- Human Immunodeficiency Virus Proteins/genetics
- Humans
- Immunization
- Macaca mulatta
- Simian Immunodeficiency Virus/physiology
- Vaccines, DNA/immunology
- Vaccines, Subunit/immunology
- Viremia
- Virus Replication
Collapse
Affiliation(s)
- Gerrit Koopman
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| | - Sam Hofman
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| | | | | | - Peter Ertl
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Phil Overend
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Cathy van Wely
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Lindy L Thomsen
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Britta Wahren
- Swedish Institute for Infectious Disease Control, Karolinska Institutet, Stockholm, Sweden
| | - Gerald Voss
- GlaxoSmithKline Biologicals, Rixensart, Belgium
| | - Jonathan L Heeney
- Department of Veterinary Medicine, University of Cambridge, UK
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| |
Collapse
|
31
|
Abstract
Synthetic peptide vaccines have potential to control viral infections. Successful experimental models using this approach include the protection of mice against the lethal Sendai virus infection by MHC class I binding CTL peptide epitope. The main benefit of vaccination with peptide epitopes is the ability to minimize the amount and complexity of a well-defined antigen. An appropriate peptide immunogen would also decrease the chance of stimulating a response against self-antigens, thereby providing a safer vaccine by avoiding autoimmunity. In general, the peptide vaccine strategy needs to dissect the specificity of antigen processing, the presence of B-and T-cell epitopes and the MHC restriction of the T-cell responses. This article briefly reviews the implications in the design of peptide vaccines and discusses the various approaches that are applied to improve their immunogenicity.
Collapse
Affiliation(s)
- Ali Azizi
- Variation Biotechnologies Inc., 22 de Varennes, Suite 210, Gatineau, QC J8T 8R1, Canada
| | | |
Collapse
|
32
|
Cristillo AD, Lisziewicz J, He L, Lori F, Galmin L, Trocio JN, Unangst T, Whitman L, Hudacik L, Bakare N, Whitney S, Restrepo S, Suschak J, Ferrari MG, Chung HK, Kalyanaraman VS, Markham P, Pal R. HIV-1 prophylactic vaccine comprised of topical DermaVir prime and protein boost elicits cellular immune responses and controls pathogenic R5 SHIV162P3. Virology 2007; 366:197-211. [PMID: 17499328 DOI: 10.1016/j.virol.2007.04.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 03/29/2007] [Accepted: 04/11/2007] [Indexed: 11/30/2022]
Abstract
Topical DNA vaccination (DermaVir) facilitates antigen presentation to naive T cells. DermaVir immunization in mice, using HIV-1 Env and Gag, elicited cellular immune responses. Boosting with HIV-1 gp120 Env and p41 Gag augmented Th1 cytokine levels. Intramuscular DNA administration was less efficient in priming antigen-specific cytokine production and memory T cells. In rhesus macaques, DermaVir immunization induced Gag- and Env-specific Th1 and Th2 cytokines and generation of memory T cells. Boosting of DermaVir-primed serum antibody levels was noted following gp140(SHIV89.6P)/p27(SIV) immunization. Rectal challenge with pathogenic R5-tropic SHIV162P3 resulted in control of plasma viremia (4/5 animals) that was reflected in jejunum, colon and mesenteric lymph nodes. An inverse correlation was found between Gag- and Env-specific central memory T cell responses on the day of challenge and plasma viremia at set point. Overall, the topical DermaVir/protein vaccination yields central memory T cell responses and facilitates control of pathogenic SHIV infection.
Collapse
Affiliation(s)
- Anthony D Cristillo
- Advanced BioScience Laboratories, Inc., 5510 Nicholson Lane, Kensington, MD 20895, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Shu Y, Winfrey S, Yang ZY, Xu L, Rao SS, Srivastava I, Barnett SW, Nabel GJ, Mascola JR. Efficient protein boosting after plasmid DNA or recombinant adenovirus immunization with HIV-1 vaccine constructs. Vaccine 2006; 25:1398-408. [PMID: 17113201 PMCID: PMC1821094 DOI: 10.1016/j.vaccine.2006.10.046] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 10/23/2006] [Accepted: 10/24/2006] [Indexed: 01/13/2023]
Abstract
DNA plasmids and recombinant adenovirus serotype-5 (rAd5) vectors are being studied in human clinical trials as HIV-1 vaccine candidates. Each elicits robust T-cell responses and modest antibody levels. Since protein immunization alone elicits antibody but not CD8 T-cell responses, we studied protein boosting of DNA and rAd5 HIV-1 vaccine vectors. A single Env protein immunization provided a marked boost in antibody titer in guinea pigs primed with either DNA or rAd5 vaccines, and the resulting antibody binding and neutralization levels were similar to those attained after thee sequential protein immunizations. Since both T-cell immunity and neutralizing antibodies are thought to be required for protection against HIV-1, it may be possible to establish a balanced T-cell and antibody response with appropriate vectored vaccines and improve the neutralizing antibody titer with protein boosting.
Collapse
Affiliation(s)
- Yuuei Shu
- Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|