45 results on '"Hannaman D"'
Search Results
2. Electroporation (EP)-related technical errors experienced during an HIV vaccine clinical trial conducted in Rwanda and Uganda: lessons learned
- Author
-
Bayingana R, Nanvubya A, Karita E, Nyombayire J, Ingabire R, Chinyenze K, Lehrman J, Schimidt C, Hannaman D, Allen S, and Fast P
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
3. OA05-01. In vivo electroporation enhances the immunogenicity of ADVAX, a DNA-based HIV-1 vaccine candidate, in healthy volunteers
- Author
-
Park H, Schmidt C, Smith C, Dally L, Clark L, Cheeseman H, Gill DK, Tarragona T, Cox J, Huang Y, Andersen J, Caskey M, Vittorino RM, Boente-Carrera MM, Dugin DP, Gardiner DF, Hannaman D, Schlesinger SJ, Hurley A, Vasan S, Sayeed E, Gilmour J, Fast P, Bernard R, and Ho DD
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2009
- Full Text
- View/download PDF
4. A Phase 1 clinical trial of Hantaan virus and Puumala virus M-segment DNA vaccines for haemorrhagic fever with renal syndrome delivered by intramuscular electroporation
- Author
-
Hooper, J.W., Moon, J.E., Paolino, K.M., Newcomer, R., McLain, D.E., Josleyn, M., Hannaman, D., and Schmaljohn, C.
- Published
- 2014
- Full Text
- View/download PDF
5. A Phase 2a Randomized, Double-Blind, Dose-Optimizing Study to Evaluate the Immunogenicity and Safety of a Bivalent DNA Vaccine for Hemorrhagic Fever with Renal Syndrome Delivered by Intramuscular Electroporation
- Author
-
Hooper, Jay, primary, Paolino, K. M., additional, Mills, K., additional, Kwilas, S., additional, Josleyn, M., additional, Cohen, M., additional, Somerville, B., additional, Wisniewski, M., additional, Norris, S., additional, Hill, B., additional, Sanchez-Lockhart, M., additional, Hannaman, D., additional, and Schmaljohn, C. S., additional
- Published
- 2020
- Full Text
- View/download PDF
6. Combined skin and muscle vaccination differentially impact the quality of effector T cell functions: the CUTHIVAC-001 randomized trial
- Author
-
Haidari, G., primary, Cope, A., additional, Miller, A., additional, Venables, S., additional, Yan, C., additional, Ridgers, H., additional, Reijonen, K., additional, Hannaman, D., additional, Spentzou, A., additional, Hayes, P., additional, Bouliotis, G., additional, Vogt, A., additional, Joseph, S., additional, Combadiere, B., additional, McCormack, S., additional, and Shattock, R. J., additional
- Published
- 2017
- Full Text
- View/download PDF
7. A clinical trial of a DNA vaccine (SCIB1) that targets dendritic cells in vivo in fully resected melanoma patients; a vaccine to prevent disease recurrence?
- Author
-
Durrant, L G, Ottensmeier, C H, Mulatero, C, Lorigan, P, Plummer, R, Cunnell, M, Metheringham, R, Brentville, V, Machado, Lee, Daniels, I, Hannaman, D, and Patel, P M
- Abstract
Background: SCIB1 is a DNA vaccine encoding a human IgG1 antibody with CDRs that contain four epitopes from two melanoma antigens (three from gp100 and one from TRP2). The vaccine elicits potent anti-tumour responses by stimulating high frequency, high avidity T-cells via both direct and cross-presentation of antibody. A clinical study in stage III/IV melanoma patients, all with tumour present at study entry, showed that 2-8mg doses could induce T-cell responses in 7/9 patients with no associated toxicity. Encouragingly overall survival was 31 months. This study addresses the question as to whether SCIB1 can be used as an adjuvant therapy in fully resected melanoma patients to prevent further disease. Methods: Sixteen patients with fully resected stage III (n=9) or stage IV (n=7) melanoma were immunised with 4mg of SCIB1 by intramuscular electroporation at 3 weekly intervals and subsequently at 3 and 6 months. Patients could continue treatment for 5 years. Results: All 16 patients showed vaccine-epitope-specific T-cell responses (i.e. proliferation ex vivo and/or γIFN Elispot responses in-vitro). Twelve patients responded to all four epitopes, two patients to three epitopes, one to two epitopes and one to a single epitope. Five patients remain in the continuation phase - all show strong T-cell memory responses following boosting. At present, median survival time is 37 months from trial entry and 41.5 months from diagnosis of metastases. Overall survival is 100% for both groups. Five patients relapsed at 1, 4, 14, 17 and 18 months but have shown no further recurrences at follow-up. Conclusion: These results show that a DNA vaccine encoding epitopes from melanoma antigens can induce measurable T-cell responses and, furthermore, it may confer protection from recurrence of melanoma with little associated toxicity. SCIB1 deserves further evaluation as an adjuvant therapy.
- Published
- 2015
8. Electroporation Enhances Immunogenicity of a DNA Vaccine Expressing Woodchuck Hepatitis Virus Surface Antigen in Woodchucks
- Author
-
Liu, K. H., primary, Ascenzi, M. A., additional, Bellezza, C. A., additional, Bezuidenhout, A. J., additional, Cote, P. J., additional, Gonzalez-Aseguinolaza, G., additional, Hannaman, D., additional, Luxembourg, A., additional, Evans, C. F., additional, Tennant, B. C., additional, and Menne, S., additional
- Published
- 2011
- Full Text
- View/download PDF
9. OA05-01. In vivo electroporation enhances the immunogenicity of ADVAX, a DNA-based HIV-1 vaccine candidate, in healthy volunteers
- Author
-
Vasan, S, primary, Hurley, A, additional, Schlesinger, SJ, additional, Hannaman, D, additional, Gardiner, DF, additional, Dugin, DP, additional, Boente-Carrera, MM, additional, Vittorino, RM, additional, Caskey, M, additional, Andersen, J, additional, Huang, Y, additional, Cox, J, additional, Tarragona, T, additional, Gill, DK, additional, Cheeseman, H, additional, Clark, L, additional, Dally, L, additional, Smith, C, additional, Schmidt, C, additional, Park, H, additional, Sayeed, E, additional, Gilmour, J, additional, Fast, P, additional, Bernard, R, additional, and Ho, DD, additional
- Published
- 2009
- Full Text
- View/download PDF
10. Specifying Battle Simulation Requirements. A Model and Case History.
- Author
-
HUMAN RESOURCES RESEARCH ORGANIZATION ALEXANDRIA VA, Hannaman,D L, HUMAN RESOURCES RESEARCH ORGANIZATION ALEXANDRIA VA, and Hannaman,D L
- Abstract
The purpose of this paper is to present a model that can be of help to military users who have to prepare functional requirements for battle simulations. The particular functional requirements are for a battle simulation that will be used to conduct research on training command, control, and communications (C3) skills among the four leaders of a tank platoon (i.e., the platoon leader, the platoon sergeant, and the two tank commanders) during the performance of combined arms operations. This battle simulation, SIMCAT (SIMulation in Combined Arms Training), is being developed for the U.S. Army Research Institute for the Behavioral and Social Sciences (ARI) by the Human Resources Research Organization (HumRRo) under contract MDA903-83C-0504 with Perceptronics as a subcontractor. The functional requirements were prepared for ARI by HumRRO and were provided to Perceptronics for the hardware/software development of SIMCAT. While these functional requirements are specific to the development of SIMCAT and the purposes for which SIMCAT was intended, they are being presented in this paper to serve as a model for others who must prepare functional requirements for battle simulations. The functions for which requirements are provided (e.g., terrain, movement, engagement, communications) must be considered in the development of any battle simulation. While the requirements will obviously vary from one simulation to nother, it is hoped that their publication will assist others who will undertake similar tasks.
- Published
- 1984
11. Development of a Computer-Assisted Simulation of Tactical Voice Communications. Phase I. Conceptual Design.
- Author
-
HUMAN RESOURCES RESEARCH ORGANIZATION ALEXANDRIA VA, Hannaman,D L, Michaelis,P R, Chambers,R M, HUMAN RESOURCES RESEARCH ORGANIZATION ALEXANDRIA VA, Hannaman,D L, Michaelis,P R, and Chambers,R M
- Abstract
The dramatic introduction of technology onto the battlefield has intensified in recent years and is expected to continue at an accelerated rate. This results in increased human performance demands. Human performance is physiologically limited in terms of the rate at which a human can process information and physically perform tasks. Though there is little that can be done about the human's physiological limitations, human performance capabilities can be improved in other ways. Human performance capabilities of Army personnel can best be improved by either raising enlistment standards (e.g., education, aptitude) and/or providing more and better training. Enlistment standards are expected to remain relatively constant during the foreseeable future. Though Army training has improved and will continue to do so, the training environment is burdened with ever increasing demands (to a large extent attributable to the proliferation of technology on the battlefield) while simultaneously experiencing diminishing resources (e.g., time and money). As a result, human performance capabilities in the Army are also expected to remain relatively constant.
- Published
- 1983
12. A Systems Definition and Evaluation of Technology Alternatives for ACES (Army Continuing Education System).
- Author
-
HUMAN RESOURCES RESEARCH ORGANIZATION ALEXANDRIA VA, Seidel,R J, Hannaman,D L, Hillelsohn,M J, Wagner,H, HUMAN RESOURCES RESEARCH ORGANIZATION ALEXANDRIA VA, Seidel,R J, Hannaman,D L, Hillelsohn,M J, and Wagner,H
- Abstract
The objectives of this report were to: define the ACES(Army Continuing Education System) system in terms of its objectives, major components, data requirements, major processes, information flow, and organizational components; identify the major deficiencies inherent in the ACES; identify current technology and R&D initiatives and the degree to which they do and/or will impact identified deficiencies; present recommendations and guidelines for the acquisition of additional technology necessary to overcome ACES deficiencies that will not be eliminated by current technology and or R&D initiatives. (Author)
- Published
- 1982
13. Focusing HIV-1 Gag T cell responses to highly conserved regions by DNA vaccination in HVTN 119.
- Author
-
Kalams SA, Felber BK, Mullins JI, Scott HM, Allen MA, De Rosa SC, Heptinstall J, Tomaras GD, Hu J, DeCamp AC, Rosati M, Bear J, Pensiero MN, Eldridge J, Egan MA, Hannaman D, McElrath MJ, and Pavlakis GN
- Subjects
- Humans, Female, Adult, Male, Double-Blind Method, Middle Aged, Young Adult, T-Lymphocytes immunology, HIV Antibodies immunology, Vaccination methods, Immunogenicity, Vaccine, CD4-Positive T-Lymphocytes immunology, Vaccines, DNA immunology, Vaccines, DNA administration & dosage, HIV-1 immunology, AIDS Vaccines immunology, AIDS Vaccines administration & dosage, HIV Infections immunology, HIV Infections prevention & control, gag Gene Products, Human Immunodeficiency Virus immunology, gag Gene Products, Human Immunodeficiency Virus genetics
- Abstract
BACKGROUNDAn HIV-1 DNA vaccine composed of 7 highly conserved, structurally important elements (conserved elements, CE) of p24Gag was tested in a phase I randomized, double-blind clinical trial (HVTN 119, NCT03181789) in people without HIV. DNA vaccination of CE prime/CE+p55Gag boost was compared with p55Gag.METHODSTwo groups (n = 25) received 4 DNA vaccinations (CE/CE+p55Gag or p55Gag) by intramuscular injection/electroporation, including IL-12 DNA adjuvant. The placebo group (n = 6) received saline. Participants were followed for safety and tolerability. Immunogenicity was assessed for T cell and antibody responses.RESULTSBoth regimens were safe and generally well tolerated. The p24CE vaccine was immunogenic and significantly boosted by CE+p55Gag (64% CD4+, P = 0.037; 42% CD8+, P = 0.004). CE+p55Gag induced responses to 5 of 7 CE, compared with only 2 CE by p55Gag DNA, with a higher response to CE5 in 30% of individuals (P = 0.006). CE+p55Gag induced significantly higher CD4+ CE T cell breadth (0.68 vs. 0.22 CE; P = 0.029) and a strong trend for overall T cell breadth (1.14 vs. 0.52 CE; P = 0.051). Both groups developed high cellular and humoral responses. p24CE vaccine-induced CD4+ CE T cell responses correlated (P = 0.007) with p24Gag antibody responses.CONCLUSIONThe CE/CE+p55Gag DNA vaccine induced T cell responses to conserved regions in p24Gag, increasing breadth and epitope recognition throughout p55Gag compared with p55Gag DNA. Vaccines focusing immune responses by priming responses to highly conserved regions could be part of a comprehensive HIV vaccine strategy.TRIAL REGISTRATIONClinical Trials.gov NCT03181789FUNDINGHVTN, NIAID/NIH.
- Published
- 2024
- Full Text
- View/download PDF
14. Acceptability and tolerability of repeated intramuscular electroporation of Multi-antigenic HIV (HIVMAG) DNA vaccine among healthy African participants in a phase 1 randomized controlled trial.
- Author
-
Mpendo J, Mutua G, Nanvubya A, Anzala O, Nyombayire J, Karita E, Dally L, Hannaman D, Price M, Fast PE, Priddy F, Gelderblom HC, and Hills NK
- Subjects
- Adolescent, Adult, Double-Blind Method, Female, Humans, Male, Middle Aged, AIDS Vaccines administration & dosage, Electroporation, Muscle Contraction, Muscle, Skeletal, Vaccination, Vaccines, DNA administration & dosage
- Abstract
Introduction: Intramuscular electroporation (IM/EP) is a vaccine delivery technique that improves the immunogenicity of DNA vaccines. We evaluated the acceptability and tolerability of electroporation among healthy African study participants., Methods: Forty-five participants were administered a DNA vaccine (HIV-MAG) or placebo by electroporation at three visits occurring at four week-intervals. At the end of each visit, participants were asked to rate pain at four times: (1) when the device was placed on the skin and vaccine injected, before the electrical stimulation, (2) at the time of electrical stimulation and muscle contraction, and (3) at 10 minutes and (4) 30 minutes after the procedure was completed. For analyses, pain level was dichotomized as either "acceptable" (none/slight/uncomfortable) or "too much" (Intense, severe, and very severe) and examined over time using repeated measures models. Optional brief comments made by participants were summarized anecdotally., Results: All 45 participants completed all three vaccination visits; none withdrew from the study due to the electroporation procedure. Most (76%) reported pain levels as acceptable at every time point across all vaccination visits. The majority of "unacceptable" pain was reported at the time of electrical stimulation. The majority of the participants (97%) commented that they preferred electroporation to standard injection., Conclusion: Repeated intramuscular electroporation for vaccine delivery was found to be acceptable and feasible among healthy African HIV vaccine trial participants. The majority of participants reported an acceptable pain level at all vaccination time points. Further investigation may be warranted into the value of EP to improve immunization outcomes. ClinicalTrials.gov NCT01496989., Competing Interests: PEF, MP and FP were employees of IAVI at the time of the study. DH is an employee of Ichor Medical Sciences Inc which owns the rights to the TriGrid Delivery System. The rest of the authors have no competing interests.
- Published
- 2020
- Full Text
- View/download PDF
15. The Safety and Immunogenicity of GTU ® MultiHIV DNA Vaccine Delivered by Transcutaneous and Intramuscular Injection With or Without Electroporation in HIV-1 Positive Subjects on Suppressive ART.
- Author
-
Haidari G, Day S, Wood M, Ridgers H, Cope AV, Fleck S, Yan C, Reijonen K, Hannaman D, Spentzou A, Hayes P, Vogt A, Combadiere B, Cook A, McCormack S, and Shattock RJ
- Subjects
- AIDS Vaccines administration & dosage, AIDS Vaccines adverse effects, Administration, Cutaneous, Antiretroviral Therapy, Highly Active, Cytokines metabolism, Electroporation, HIV Infections drug therapy, Humans, Injections, Intramuscular, Patient Outcome Assessment, Vaccination, Vaccines, DNA administration & dosage, Vaccines, DNA adverse effects, AIDS Vaccines immunology, HIV Infections immunology, HIV Infections prevention & control, HIV-1 immunology, Immunogenicity, Vaccine, Vaccines, DNA immunology
- Abstract
Previous studies have shown targeting different tissues via the transcutaneous (TC) and intramuscular injection (IM) with or without electroporation (EP) has the potential to trigger immune responses to DNA vaccination. The CUTHIVTHER 001 Phase I/II randomized controlled clinical trial was designed to determine whether the mode of DNA vaccination delivery (TC+IM or EP+IM) could influence the quality and function of induced cellular immune responses compared to placebo, in an HIV positive clade B cohort on antiretroviral therapy (ART). The GTU
® MultiHIV B DNA vaccine DNA vaccine encoded a MultiHIV B clade fusion protein to target the cellular response. Overall the vaccine and regimens were safe and well-tolerated. There were robust pre-vaccination IFN-γ responses with no measurable change following vaccination compared to placebo. However, modest intracellular cytokine staining (ICS) responses were seen in the TC+IM group. A high proportion of individuals demonstrated potent viral inhibition at baseline that was not improved by vaccination. These results show that HIV positive subjects with nadir CD4+ counts ≥250 on suppressive ART display potent levels of cellular immunity and viral inhibition, and that DNA vaccination alone is insufficient to improve such responses. These data suggest that more potent prime-boost vaccination strategies are likely needed to improve pre-existing responses in similar HIV-1 cohorts (This study has been registered at http://ClinicalTrials.gov under registration no. NCT02457689)., (Copyright © 2019 Haidari, Day, Wood, Ridgers, Cope, Fleck, Yan, Reijonen, Hannaman, Spentzou, Hayes, Vogt, Combadiere, Cook, McCormack and Shattock.)- Published
- 2019
- Full Text
- View/download PDF
16. Glycosylation of HIV Env Impacts IgG Subtype Responses to Vaccination.
- Author
-
Heß R, Storcksdieck Genannt Bonsmann M, Lapuente D, Maaske A, Kirschning C, Ruland J, Lepenies B, Hannaman D, Tenbusch M, and Überla K
- Subjects
- Animals, Glycosylation, Hemagglutinin Glycoproteins, Influenza Virus genetics, Hemagglutinin Glycoproteins, Influenza Virus immunology, Immunity, Humoral, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Th2 Cells immunology, Viral Fusion Proteins genetics, Viral Fusion Proteins immunology, AIDS Vaccines immunology, HIV Antibodies immunology, Immunoglobulin G immunology, Vaccines, DNA immunology, env Gene Products, Human Immunodeficiency Virus immunology
- Abstract
The envelope protein (Env) is the only surface protein of the human immunodeficiency virus (HIV) and as such the exclusive target for protective antibody responses. Experimental evidences from mouse models suggest a modulating property of Env to steer antibody class switching towards the less effective antibody subclass IgG1 accompanied with strong TH2 helper responses. By simple physical linkage we were able to imprint this bias, exemplified by a low IgG2a/IgG1 ratio of antigen-specific antibodies, onto an unrelated antigen, namely the HIV capsid protein p24. Here, our results indicate the glycan moiety of Env as the responsible immune modulating activity. Firstly, in Card9
-/- mice lacking specific C-Type lectin responsiveness, DNA immunization significantly increased the IgG2a/IgG1 ratio for the Env-specific antibodies while the antibody response against the F-protein of the respiratory syncytial virus (RSV) serving as control antigen remained unchanged. Secondly, sequential shortening of the Env encoding sequence revealed the C2V3 domain as responsible for the strong IgG1 responses and TH2 cytokine production. Removing all potential N-glycosylation sites from the C2V3 domain by site-specific mutagenesis reversed the vaccine-induced immune response towards a Th1-dominated T-cell response and a balanced IgG2a/IgG1 ratio. Accordingly, the stretch of oligomannose glycans in the C2V3 domain of Env might mediate a specific uptake and/or signaling modus in antigen presenting cells by involving interaction with an as yet unknown C-type lectin receptor. Our results contribute to a deeper understanding of the impact of Env glycosylation on HIV antigen-specific immune responses, which will further support HIV vaccine development.- Published
- 2019
- Full Text
- View/download PDF
17. Safety and tolerability of HIV-1 multiantigen pDNA vaccine given with IL-12 plasmid DNA via electroporation, boosted with a recombinant vesicular stomatitis virus HIV Gag vaccine in healthy volunteers in a randomized, controlled clinical trial.
- Author
-
Elizaga ML, Li SS, Kochar NK, Wilson GJ, Allen MA, Tieu HVN, Frank I, Sobieszczyk ME, Cohen KW, Sanchez B, Latham TE, Clarke DK, Egan MA, Eldridge JH, Hannaman D, Xu R, Ota-Setlik A, McElrath MJ, and Hay CM
- Subjects
- AIDS Vaccines adverse effects, Adult, Combined Modality Therapy, Double-Blind Method, Electroporation, Female, Genetic Vectors adverse effects, HIV-1, Healthy Volunteers, Humans, Immunization, Secondary, Injections, Intramuscular, Male, Middle Aged, Plasmids genetics, Vaccines, Attenuated adverse effects, Vaccines, DNA adverse effects, Young Adult, gag Gene Products, Human Immunodeficiency Virus, AIDS Vaccines administration & dosage, Genetic Vectors administration & dosage, Interleukin-12 genetics, Vaccines, Attenuated administration & dosage, Vaccines, DNA administration & dosage, Vesicular stomatitis Indiana virus genetics
- Abstract
Background: The addition of plasmid cytokine adjuvants, electroporation, and live attenuated viral vectors may further optimize immune responses to DNA vaccines in heterologous prime-boost combinations. The objective of this study was to test the safety and tolerability of a novel prime-boost vaccine regimen incorporating these strategies with different doses of IL-12 plasmid DNA adjuvant., Methods: In a phase 1 study, 88 participants received an HIV-1 multiantigen (gag/pol, env, nef/tat/vif) DNA vaccine (HIV-MAG, 3000 μg) co-administered with IL-12 plasmid DNA adjuvant at 0, 250, 1000, or 1500 μg (N = 22/group) given intramuscularly with electroporation (Ichor TriGrid™ Delivery System device) at 0, 1 and 3 months; followed by attenuated recombinant vesicular stomatitis virus, serotype Indiana, expressing HIV-1 Gag (VSV-Gag), 3.4 ⊆ 107 plaque-forming units (PFU), at 6 months; 12 others received placebo. Injections were in both deltoids at each timepoint. Participants were monitored for safety and tolerability for 15 months., Results: The dose of IL-12 pDNA did not increase pain scores, reactogenicity, or adverse events with the co-administered DNA vaccine, or following the VSV-Gag boost. Injection site pain and reactogenicity were common with intramuscular injections with electroporation, but acceptable to most participants. VSV-Gag vaccine often caused systemic reactogenicity symptoms, including a viral syndrome (in 41%) of fever, chills, malaise/fatigue, myalgia, and headache; and decreased lymphocyte counts 1 day after vaccination., Conclusions: HIV-MAG DNA vaccine given by intramuscular injection with electroporation was safe at all doses of IL-12 pDNA. The VSV-Gag vaccine at this dose was associated with fever and viral symptoms in some participants, but the vaccine regimens were safe and generally well-tolerated., Trial Registration: Clinical Trials.gov NCT01578889., Competing Interests: The authors have read the journal policy and the authors of this manuscript have the following competing interests: MAA is employed by the National Institute of Allergy and Infectious Diseases (NIAID), the study sponsor. MLE, SSL, NKK, GJW, HVNT, IF, MES, KWC, BSP, JHE, MJM and CMH are recipients of NIAID funding, and this publication is a result of activities funded by NIAID. MAA is not involved with the process of funding these awards, nor in their administration of scientific aspects and, in accordance with NIAID policies, is deferred from decisions regarding funding of coauthors for a requisite period. TEL, DKC, JHE, RX, and AOS are employees and stakeholders of Profectus BioSciences, Inc. DKC is the principal author/inventor on a patent for the rVSVN4CT1 vector. At the time the study was conducted, MAE was an employee and stakeholder of Profectus BioSciences, Inc. and is now an employee of Takeda Pharmaceuticals, which has no affiliation with the study. DH is an employee and stakeholder of Ichor Medical Systems, Inc. IF serves on advisory boards for Gilead Sciences and ViiV Healthcare. HVNT has received research grant funding from Merck & Co. These affiliations do not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2018
- Full Text
- View/download PDF
18. Combined Skin and Muscle DNA Priming Provides Enhanced Humoral Responses to a Human Immunodeficency Virus Type 1 Clade C Envelope Vaccine.
- Author
-
Cheeseman HM, Day S, McFarlane LR, Fleck S, Miller A, Cole T, Sousa-Santos N, Cope A, Cizmeci D, Tolazzi M, Hwekwete E, Hannaman D, Kratochvil S, McKay PF, Chung AW, Kent SJ, Cook A, Scarlatti G, Abraham S, Combadiere B, McCormack S, Lewis DJ, and Shattock RJ
- Subjects
- AIDS Vaccines administration & dosage, AIDS Vaccines immunology, Adolescent, Adult, Animals, CD4-Positive T-Lymphocytes drug effects, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, DNA Primers immunology, Electroporation, Female, HIV-1 immunology, HIV-1 pathogenicity, Healthy Volunteers, Histocompatibility Antigens Class II genetics, Histocompatibility Antigens Class II immunology, Humans, Immunity, Cellular drug effects, Immunity, Cellular immunology, Immunity, Humoral drug effects, Immunity, Humoral immunology, Injections, Intradermal, Injections, Intramuscular, Male, Middle Aged, Vaccination methods, Vaccines, DNA immunology, Young Adult, env Gene Products, Human Immunodeficiency Virus genetics, env Gene Products, Human Immunodeficiency Virus immunology, Antibodies, Neutralizing immunology, HIV Antibodies immunology, Vaccines, DNA administration & dosage, env Gene Products, Human Immunodeficiency Virus administration & dosage
- Abstract
Intradermal (i.d.) and intramuscular (i.m.) injections when administered with or without electroporation (EP) have the potential to tailor the immune response to DNA vaccination. This Phase I randomized controlled clinical trial in human immunodeficiency virus type 1-negative volunteers investigated whether the site and mode of DNA vaccination influences the quality of induced cellular and humoral immune responses following the DNA priming phase and subsequent protein boost with recombinant clade C CN54 gp140. A strategy of concurrent i.d. and i.m. DNA immunizations administered with or without EP was adopted. Subtle differences were observed in the shaping of vaccine-induced virus-specific CD4+ and CD8+ T cell-mediated immune responses between groups receiving: i.d.
EP + i.m., i.d. + i.m.EP , and i.d.EP + i.m.EP regimens. The DNA priming phase induced 100% seroconversion in all of the groups. A single, non-adjuvanted protein boost induced a rapid and profound increase in binding antibodies in all groups, with a trend for higher responses in i.d.EP + i.m.EP . The magnitude of antigen-specific binding immunoglobulin G correlated with neutralization of closely matched clade C 93MW965 virus and Fc-dimer receptor binding (FcγRIIa and FcγRIIIa). These results offer new perspectives on the use of combined skin and muscle DNA immunization in priming humoral and cellular responses to recombinant protein.- Published
- 2018
- Full Text
- View/download PDF
19. A Multiagent Alphavirus DNA Vaccine Delivered by Intramuscular Electroporation Elicits Robust and Durable Virus-Specific Immune Responses in Mice and Rabbits and Completely Protects Mice against Lethal Venezuelan, Western, and Eastern Equine Encephalitis Virus Aerosol Challenges.
- Author
-
Dupuy LC, Richards MJ, Livingston BD, Hannaman D, and Schmaljohn CS
- Subjects
- Aerosols, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Antibody Specificity immunology, Disease Models, Animal, Electroporation, Female, Immunity, Cellular immunology, Immunization, Mice, Rabbits, Vaccines, DNA administration & dosage, Viral Vaccines administration & dosage, Alphavirus genetics, Alphavirus immunology, Encephalitis Virus, Eastern Equine immunology, Encephalomyelitis, Equine immunology, Encephalomyelitis, Equine prevention & control, Genetic Vectors administration & dosage, Genetic Vectors genetics, Genetic Vectors immunology, Vaccines, DNA immunology, Viral Vaccines immunology
- Abstract
There remains a need for vaccines that can safely and effectively protect against the biological threat agents Venezuelan (VEEV), western (WEEV), and eastern (EEEV) equine encephalitis virus. Previously, we demonstrated that a VEEV DNA vaccine that was optimized for increased antigen expression and delivered by intramuscular (IM) electroporation (EP) elicited robust and durable virus-specific antibody responses in multiple animal species and provided complete protection against VEEV aerosol challenge in mice and nonhuman primates. Here, we performed a comparative evaluation of the immunogenicity and protective efficacy of individual optimized VEEV, WEEV, and EEEV DNA vaccines with that of a 1 : 1 : 1 mixture of these vaccines, which we have termed the 3-EEV DNA vaccine, when delivered by IM EP. The individual DNA vaccines and the 3-EEV DNA vaccine elicited robust and durable virus-specific antibody responses in mice and rabbits and completely protected mice from homologous VEEV, WEEV, and EEEV aerosol challenges. Taken together, the results from these studies demonstrate that the individual VEEV, WEEV, and EEEV DNA vaccines and the 3-EEV DNA vaccine delivered by IM EP provide an effective means of eliciting protection against lethal encephalitic alphavirus infections in a murine model and represent viable next-generation vaccine candidates that warrant further development.
- Published
- 2018
- Full Text
- View/download PDF
20. Targeting gp100 and TRP-2 with a DNA vaccine: Incorporating T cell epitopes with a human IgG1 antibody induces potent T cell responses that are associated with favourable clinical outcome in a phase I/II trial.
- Author
-
Patel PM, Ottensmeier CH, Mulatero C, Lorigan P, Plummer R, Pandha H, Elsheikh S, Hadjimichael E, Villasanti N, Adams SE, Cunnell M, Metheringham RL, Brentville VA, Machado L, Daniels I, Gijon M, Hannaman D, and Durrant LG
- Abstract
A DNA vaccine, SCIB1, incorporating two CD8 and two CD4 epitopes from TRP-2/gp100 was evaluated in patients with metastatic melanoma. Each patient received SCIB1 via intramuscular injection with electroporation. The trial was designed to find the safest dose of SCIB1 which induced immune/clinical responses in patients with or without tumour. Fifteen patients with tumor received SCIB1 doses of 0.4-8 mg whilst 20 fully-resected patients received 2-8 mg doses. Twelve patients elected to continue immunization every 3 months for up to 39 months. SCIB1 induced dose-dependent T cell responses in 88% of patients with no serious adverse effects or dose limiting toxicities. The intensity of the T cell responses was significantly higher in patients receiving 4 mg doses without tumor when compared to those with tumor ( p < 0.01). In contrast, patients with tumor showed a significantly higher response to the 8 mg dose than the 4 mg dose ( p < 0.03) but there was no significant difference in the patients without tumor. One of 15 patients with measurable disease showed an objective tumor response and 7/15 showed stable disease. 5/20 fully-resected patients have experienced disease recurrence but all remained alive at the cut-off date with a median observation time of 37 months. A positive clinical outcome was associated with MHC-I and MHC-II expression on tumors prior to therapy ( p = 0.027). We conclude that SCIB1 is well tolerated and stimulates potent T cell responses in melanoma patients. It deserves further evaluation as a single agent adjuvant therapy or in combination with checkpoint inhibitors in advanced disease.
- Published
- 2018
- Full Text
- View/download PDF
21. An immunoinformatics-derived DNA vaccine encoding human class II T cell epitopes of Ebola virus, Sudan virus, and Venezuelan equine encephalitis virus is immunogenic in HLA transgenic mice.
- Author
-
Bounds CE, Terry FE, Moise L, Hannaman D, Martin WD, De Groot AS, Suschak JJ, Dupuy LC, and Schmaljohn CS
- Subjects
- Animals, Ebolavirus genetics, Encephalitis Virus, Venezuelan Equine genetics, Enzyme-Linked Immunospot Assay, Epitopes, T-Lymphocyte genetics, Female, Histocompatibility Antigens Class II genetics, Humans, Interferon-gamma metabolism, Mice, Inbred BALB C, Mice, Transgenic, Protein Binding, T-Lymphocytes immunology, Vaccines, DNA administration & dosage, Vaccines, DNA genetics, Viral Vaccines administration & dosage, Viral Vaccines genetics, Ebolavirus immunology, Encephalitis Virus, Venezuelan Equine immunology, Epitopes, T-Lymphocyte immunology, Histocompatibility Antigens Class II metabolism, Vaccines, DNA immunology, Viral Vaccines immunology
- Abstract
Immunoinformatics tools were used to predict human leukocyte antigen (HLA) class II-restricted T cell epitopes within the envelope glycoproteins and nucleocapsid proteins of Ebola virus (EBOV) and Sudan virus (SUDV) and the structural proteins of Venezuelan equine encephalitis virus (VEEV). Selected epitopes were tested for binding to soluble HLA molecules representing 5 class II alleles (DRB1*0101, DRB1*0301, DRB1*0401, DRB1*0701, and DRB1*1501). All but one of the 25 tested peptides bound to at least one of the DRB1 alleles, and 4 of the peptides bound at least moderately or weakly to all 5 DRB1 alleles. Additional algorithms were used to design a single "string-of-beads" expression construct with 44 selected epitopes arranged to avoid creation of spurious junctional epitopes. Seventeen of these 44 predicted epitopes were conserved between the major histocompatibility complex (MHC) of humans and mice, allowing initial testing in mice. BALB/c mice vaccinated with the multi-epitope construct developed statistically significant cellular immune responses to EBOV, SUDV, and VEEV peptides as measured by interferon (IFN)-γ ELISpot assays. Significant levels of antibodies to VEEV, but not EBOV, were also detected in vaccinated BALB/c mice. To assess immunogenicity in the context of a human MHC, HLA-DR3 transgenic mice were vaccinated with the multi-epitope construct and boosted with a mixture of the 25 peptides used in the binding assays. The vaccinated HLA-DR3 mice developed significant cellular immune responses to 4 of the 25 (16%) tested individual class II peptides as measured by IFN-γ ELISpot assays. In addition, these mice developed antibodies against EBOV and VEEV as measured by ELISA. While a low but significant level of protection was observed in vaccinated transgenic mice after aerosol exposure to VEEV, no protection was observed after intraperitoneal challenge with mouse-adapted EBOV. These studies provide proof of concept for the use of an informatics approach to design a multi-agent, multi-epitope immunogen and provide a basis for further testing aimed at focusing immune responses toward desired protective T cell epitopes.
- Published
- 2017
- Full Text
- View/download PDF
22. DNA Priming Increases Frequency of T-Cell Responses to a Vesicular Stomatitis Virus HIV Vaccine with Specific Enhancement of CD8 + T-Cell Responses by Interleukin-12 Plasmid DNA.
- Author
-
Li SS, Kochar NK, Elizaga M, Hay CM, Wilson GJ, Cohen KW, De Rosa SC, Xu R, Ota-Setlik A, Morris D, Finak G, Allen M, Tieu HV, Frank I, Sobieszczyk ME, Hannaman D, Gottardo R, Gilbert PB, Tomaras GD, Corey L, Clarke DK, Egan MA, Eldridge JH, McElrath MJ, and Frahm N
- Subjects
- AIDS Vaccines administration & dosage, Adjuvants, Immunologic, Adult, Epitope Mapping, Female, Genetic Vectors, HIV Infections immunology, HIV Infections prevention & control, HIV-1 immunology, Humans, Immunization, Secondary, Interleukin-12 genetics, Male, Middle Aged, Plasmids, Vaccination, Vaccines, DNA administration & dosage, Vesicular stomatitis Indiana virus immunology, Young Adult, gag Gene Products, Human Immunodeficiency Virus genetics, gag Gene Products, Human Immunodeficiency Virus immunology, AIDS Vaccines immunology, CD8-Positive T-Lymphocytes immunology, Immunogenicity, Vaccine, Interleukin-12 immunology, Vaccines, DNA immunology, Vesicular stomatitis Indiana virus genetics
- Abstract
The HIV Vaccine Trials Network (HVTN) 087 vaccine trial assessed the effect of increasing doses of pIL-12 (interleukin-12 delivered as plasmid DNA) adjuvant on the immunogenicity of an HIV-1 multiantigen (MAG) DNA vaccine delivered by electroporation and boosted with a vaccine comprising an attenuated vesicular stomatitis virus expressing HIV-1 Gag (VSV-Gag). We randomized 100 healthy adults to receive placebo or 3 mg HIV-MAG DNA vaccine (ProfectusVax HIV-1 gag / pol or ProfectusVax nef / tat / vif , env ) coadministered with pIL-12 at 0, 250, 1,000, or 1,500 μg intramuscularly by electroporation at 0, 1, and 3 months followed by intramuscular inoculation with 3.4 × 10
7 PFU VSV-Gag vaccine at 6 months. Immune responses were assessed after the prime and boost and 6 months after the last vaccination. High-dose pIL-12 increased the magnitude of CD8+ T-cell responses postboost compared to no pIL-12 ( P = 0.02), while CD4+ T-cell responses after the prime were higher in the absence of pIL-12 than with low- and medium-dose pIL-12 ( P ≤ 0.05). The VSV boost increased Gag-specific CD4+ and CD8+ T-cell responses in all groups ( P < 0.001 for CD4+ T cells), inducing a median of four Gag epitopes in responders. Six to 9 months after the boost, responses decreased in magnitude, but CD8+ T-cell response rates were maintained. The addition of a DNA prime dramatically improved responses to the VSV vaccine tested previously in the HVTN 090 trial, leading to broad epitope targeting and maintained CD8+ T-cell response rates at early memory. The addition of high-dose pIL-12 given with a DNA prime by electroporation and boosted with VSV-Gag increased the CD8+ T-cell responses but decreased the CD4+ responses. This approach may be advantageous in reshaping the T-cell responses to a variety of chronic infections or tumors. (This study has been registered at ClinicalTrials.gov under registration no. NCT01578889.)., (Copyright © 2017 American Society for Microbiology.)- Published
- 2017
- Full Text
- View/download PDF
23. A DNA vaccine for Crimean-Congo hemorrhagic fever protects against disease and death in two lethal mouse models.
- Author
-
Garrison AR, Shoemaker CJ, Golden JW, Fitzpatrick CJ, Suschak JJ, Richards MJ, Badger CV, Six CM, Martin JD, Hannaman D, Zivcec M, Bergeron E, Koehler JW, and Schmaljohn CS
- Subjects
- Animals, Disease Models, Animal, Glycoproteins genetics, Glycoproteins immunology, Hemorrhagic Fever Virus, Crimean-Congo genetics, Hemorrhagic Fever Virus, Crimean-Congo isolation & purification, Hemorrhagic Fever, Crimean immunology, Hemorrhagic Fever, Crimean virology, Humans, Immunity, Humoral, Immunocompromised Host, Mice, Mice, Knockout, Receptor, Interferon alpha-beta deficiency, Receptor, Interferon alpha-beta genetics, Th1 Cells immunology, Th2 Cells immunology, Vaccination, Vaccines, DNA administration & dosage, Viral Proteins genetics, Viral Proteins immunology, Viral Vaccines administration & dosage, Antibodies, Neutralizing blood, Antibodies, Viral blood, Hemorrhagic Fever Virus, Crimean-Congo immunology, Hemorrhagic Fever, Crimean prevention & control, Immunogenicity, Vaccine, Vaccines, DNA immunology, Viral Vaccines immunology
- Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus capable of causing a severe hemorrhagic fever disease in humans. There are currently no licensed vaccines to prevent CCHFV-associated disease. We developed a DNA vaccine expressing the M-segment glycoprotein precursor gene of CCHFV and assessed its immunogenicity and protective efficacy in two lethal mouse models of disease: type I interferon receptor knockout (IFNAR-/-) mice; and a novel transiently immune suppressed (IS) mouse model. Vaccination of mice by muscle electroporation of the M-segment DNA vaccine elicited strong antigen-specific humoral immune responses with neutralizing titers after three vaccinations in both IFNAR-/- and IS mouse models. To compare the protective efficacy of the vaccine in the two models, groups of vaccinated mice (7-10 per group) were intraperitoneally (IP) challenged with a lethal dose of CCHFV strain IbAr 10200. Weight loss was markedly reduced in CCHFV DNA-vaccinated mice as compared to controls. Furthermore, whereas all vector-control vaccinated mice succumbed to disease by day 5, the DNA vaccine protected >60% of the animals from lethal disease. Mice from both models developed comparable levels of antibodies, but the IS mice had a more balanced Th1/Th2 response to vaccination. There were no statistical differences in the protective efficacies of the vaccine in the two models. Our results provide the first comparison of these two mouse models for assessing a vaccine against CCHFV and offer supportive data indicating that a DNA vaccine expressing the glycoprotein genes of CCHFV elicits protective immunity against CCHFV.
- Published
- 2017
- Full Text
- View/download PDF
24. Broad HIV-1 inhibition in vitro by vaccine-elicited CD8(+) T cells in African adults.
- Author
-
Mutua G, Farah B, Langat R, Indangasi J, Ogola S, Onsembe B, Kopycinski JT, Hayes P, Borthwick NJ, Ashraf A, Dally L, Barin B, Tillander A, Gilmour J, De Bont J, Crook A, Hannaman D, Cox JH, Anzala O, Fast PE, Reilly M, Chinyenze K, Jaoko W, Hanke T, and Hiv-Core 004 Study Group T
- Abstract
We are developing a pan-clade HIV-1 T-cell vaccine HIVconsv, which could complement Env vaccines for prophylaxis and be a key to HIV cure. Our strategy focuses vaccine-elicited effector T-cells on functionally and structurally conserved regions (not full-length proteins and not only epitopes) of the HIV-1 proteome, which are common to most global variants and which, if mutated, cause a replicative fitness loss. Our first clinical trial in low risk HIV-1-negative adults in Oxford demonstrated the principle that naturally mostly subdominant epitopes, when taken out of the context of full-length proteins/virus and delivered by potent regimens involving combinations of simian adenovirus and poxvirus modified vaccinia virus Ankara, can induce robust CD8(+) T cells of broad specificities and functions capable of inhibiting in vitro HIV-1 replication. Here and for the first time, we tested this strategy in low risk HIV-1-negative adults in Africa. We showed that the vaccines were well tolerated and induced high frequencies of broadly HIVconsv-specific plurifunctional T cells, which inhibited in vitro viruses from four major clades A, B, C, and D. Because sub-Saharan Africa is globally the region most affected by HIV-1/AIDS, trial HIV-CORE 004 represents an important stage in the path toward efficacy evaluation of this highly rational and promising vaccine strategy.
- Published
- 2016
- Full Text
- View/download PDF
25. Human Polyclonal Antibodies Produced through DNA Vaccination of Transchromosomal Cattle Provide Mice with Post-Exposure Protection against Lethal Zaire and Sudan Ebolaviruses.
- Author
-
Bounds CE, Kwilas SA, Kuehne AI, Brannan JM, Bakken RR, Dye JM, Hooper JW, Dupuy LC, Ellefsen B, Hannaman D, Wu H, Jiao JA, Sullivan EJ, and Schmaljohn CS
- Subjects
- Animals, Animals, Genetically Modified, Antibodies, Neutralizing immunology, Cattle genetics, Cattle immunology, Chromosomes, Artificial, Human genetics, Democratic Republic of the Congo, Ebola Vaccines immunology, Female, Hemorrhagic Fever, Ebola virology, Humans, Mice, Mice, Inbred BALB C, Mice, Knockout, Receptor, Interferon alpha-beta genetics, Sudan, Vaccination methods, Antibodies, Viral metabolism, Ebolavirus immunology, Hemorrhagic Fever, Ebola prevention & control, Post-Exposure Prophylaxis methods, Vaccines, DNA immunology
- Abstract
DNA vaccination of transchromosomal bovines (TcBs) with DNA vaccines expressing the codon-optimized (co) glycoprotein (GP) genes of Ebola virus (EBOV) and Sudan virus (SUDV) produce fully human polyclonal antibodies (pAbs) that recognize both viruses and demonstrate robust neutralizing activity. Each TcB was vaccinated by intramuscular electroporation (IM-EP) a total of four times and at each administration received 10 mg of the EBOV-GPco DNA vaccine and 10 mg of the SUDV-GPco DNA vaccine at two sites on the left and right sides, respectively. After two vaccinations, robust antibody responses (titers > 1000) were detected by ELISA against whole irradiated EBOV or SUDV and recombinant EBOV-GP or SUDV-GP (rGP) antigens, with higher titers observed for the rGP antigens. Strong, virus neutralizing antibody responses (titers >1000) were detected after three vaccinations when measured by vesicular stomatitis virus-based pseudovirion neutralization assay (PsVNA). Maximal neutralizing antibody responses were identified by traditional plaque reduction neutralization tests (PRNT) after four vaccinations. Neutralizing activity of human immunoglobulins (IgG) purified from TcB plasma collected after three vaccinations and injected intraperitoneally (IP) into mice at a 100 mg/kg dose was detected in the serum by PsVNA up to 14 days after administration. Passive transfer by IP injection of the purified IgG (100 mg/kg) to groups of BALB/c mice one day after IP challenge with mouse adapted (ma) EBOV resulted in 80% protection while all mice treated with non-specific pAbs succumbed. Similarly, interferon receptor 1 knockout (IFNAR(-/-)) mice receiving the purified IgG (100 mg/kg) by IP injection one day after IP challenge with wild type SUDV resulted in 89% survival. These results are the first to demonstrate that filovirus GP DNA vaccines administered to TcBs by IM-EP can elicit neutralizing antibodies that provide post-exposure protection. Additionally, these data describe production of fully human IgG in a large animal system, a system which is capable of producing large quantities of a clinical grade therapeutic product.
- Published
- 2015
- Full Text
- View/download PDF
26. Evaluation of the Impact of Codon Optimization and N-Linked Glycosylation on Functional Immunogenicity of Pfs25 DNA Vaccines Delivered by In Vivo Electroporation in Preclinical Studies in Mice.
- Author
-
Datta D, Bansal GP, Kumar R, Ellefsen B, Hannaman D, and Kumar N
- Subjects
- Animals, Antibodies, Protozoan blood, Antibodies, Protozoan immunology, Drug Evaluation, Preclinical, Enzyme-Linked Immunosorbent Assay, Female, Glycosylation, Malaria prevention & control, Malaria Vaccines administration & dosage, Mice, Inbred BALB C, Plasmodium berghei immunology, Plasmodium berghei pathogenicity, Protozoan Proteins genetics, Vaccines, DNA administration & dosage, Codon, Electroporation, Malaria Vaccines immunology, Protozoan Proteins immunology, Vaccines, DNA immunology
- Abstract
Plasmodium falciparum sexual stage surface antigen Pfs25 is a well-established candidate for malaria transmission-blocking vaccine development. Immunization with DNA vaccines encoding Pfs25 has been shown to elicit potent antibody responses in mice and nonhuman primates. Studies aimed at further optimization have revealed improved immunogenicity through the application of in vivo electroporation and by using a heterologous prime-boost approach. The goal of the studies reported here was to systematically evaluate the impact of codon optimization, in vivo electroporation, and N-linked glycosylation on the immunogenicity of Pfs25 encoded by DNA vaccines. The results from this study demonstrate that while codon optimization and in vivo electroporation greatly improved functional immunogenicity of Pfs25 DNA vaccines, the presence or absence of N-linked glycosylation did not significantly impact vaccine efficacy. These findings suggest that N-glycosylation of Pfs25 encoded by DNA vaccines is not detrimental to overall transmission-blocking efficacy., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
27. A Phase I Double Blind, Placebo-Controlled, Randomized Study of the Safety and Immunogenicity of Electroporated HIV DNA with or without Interleukin 12 in Prime-Boost Combinations with an Ad35 HIV Vaccine in Healthy HIV-Seronegative African Adults.
- Author
-
Mpendo J, Mutua G, Nyombayire J, Ingabire R, Nanvubya A, Anzala O, Karita E, Hayes P, Kopycinski J, Dally L, Hannaman D, Egan MA, Eldridge JH, Syvertsen K, Lehrman J, Rasmussen B, Gilmour J, Cox JH, Fast PE, and Schmidt C
- Subjects
- AIDS Vaccines immunology, Adenoviridae metabolism, Adult, CD8-Positive T-Lymphocytes immunology, Demography, Double-Blind Method, Enzyme-Linked Immunospot Assay, Female, Flow Cytometry, HIV Antibodies immunology, Humans, Immunity, Cellular, Immunity, Humoral, Immunization, Interferon-gamma metabolism, Male, Middle Aged, Placebos, Young Adult, AIDS Vaccines adverse effects, DNA, Viral adverse effects, DNA, Viral immunology, Electroporation, HIV Infections immunology, Immunization, Secondary, Interleukin-12 immunology
- Abstract
Background: Strategies to enhance the immunogenicity of DNA vaccines in humans include i) co-administration of molecular adjuvants, ii) intramuscular administration followed by in vivo electroporation (IM/EP) and/or iii) boosting with a different vaccine. Combining these strategies provided protection of macaques challenged with SIV; this clinical trial was designed to mimic the vaccine regimen in the SIV study., Methods: Seventy five healthy, HIV-seronegative adults were enrolled into a phase 1, randomized, double-blind, placebo-controlled trial. Multi-antigenic HIV (HIVMAG) plasmid DNA (pDNA) vaccine alone or co-administered with pDNA encoding human Interleukin 12 (IL-12) (GENEVAX IL-12) given by IM/EP using the TriGrid Delivery System was tested in different prime-boost regimens with recombinant Ad35 HIV vaccine given IM., Results: All local reactions but one were mild or moderate. Systemic reactions and unsolicited adverse events including laboratory abnormalities did not differ between vaccine and placebo recipients. No serious adverse events (SAEs) were reported. T cell and antibody response rates after HIVMAG (x3) prime-Ad35 (x1) boost were independent of IL-12, while the magnitude of interferon gamma (IFN-γ) ELISPOT responses was highest after HIVMAG (x3) without IL-12. The quality and phenotype of T cell responses shown by intracellular cytokine staining (ICS) were similar between groups. Inhibition of HIV replication by autologous T cells was demonstrated after HIVMAG (x3) prime and was boosted after Ad35. HIV specific antibodies were detected only after Ad35 boost, although there was a priming effect with 3 doses of HIVMAG with or without IL-12. No anti-IL-12 antibodies were detected., Conclusion: The vaccines were safe, well tolerated and moderately immunogenic. Repeated administration IM/EP was well accepted. An adjuvant effect of co-administered plasmid IL-12 was not detected., Trial Registration: ClinicalTrials.gov NCT01496989.
- Published
- 2015
- Full Text
- View/download PDF
28. DNA vaccines encoding DEC205-targeted antigens: immunity or tolerance?
- Author
-
Niezold T, Storcksdieck Genannt Bonsmann M, Maaske A, Temchura V, Heinecke V, Hannaman D, Buer J, Ehrhardt C, Hansen W, Überla K, and Tenbusch M
- Subjects
- Animals, Antigens, Viral genetics, Antigens, Viral immunology, Antigens, Viral pharmacology, Asthma immunology, Asthma prevention & control, Female, HEK293 Cells, Histocompatibility Antigens Class II immunology, Humans, Influenza A virus genetics, Influenza Vaccines genetics, Influenza Vaccines immunology, Mice, Minor Histocompatibility Antigens, Orthomyxoviridae Infections prevention & control, Vaccines, DNA genetics, Vaccines, DNA immunology, Antigens, CD immunology, CD4-Positive T-Lymphocytes immunology, Immune Tolerance drug effects, Immunity, Cellular drug effects, Influenza A virus immunology, Influenza Vaccines pharmacology, Lectins, C-Type immunology, Orthomyxoviridae Infections immunology, Receptors, Cell Surface immunology, Vaccines, DNA pharmacology
- Abstract
Targeting of antigens to the endocytic uptake receptor DEC205 resulted in enhanced antigen presentation by dendritic cells (DCs). In combination with adjuvants for DC maturation, proteins coupled to an antibody against DEC205 induced strong pathogen-specific immune responses, whereas without additional adjuvant tolerance could be induced. As less is known about DNA vaccines encoding DEC205-targeted antigens, we explored the immunogenicity and efficacy of a dendritic cell-targeted DNA vaccine against influenza A virus (IAV) delivered by electroporation. Although coupling of haemagglutinin to a single-chain antibody against DEC205 enhanced antigen presentation on MHC class II and activation of T-cell receptor-transgenic CD4 T cells, the T-cell responses induced by the targeted DNA vaccine in wild-type BALB/c mice were significantly reduced compared with DNA encoding non-targeted antigens. Consistently, these mice were less protected against an IAV infection. Adoptive transfer experiments were performed to assess the fate of the antigen-specific T cells in animals vaccinated with DNA encoding DEC205-targeted antigens. By this, we could exclude the general deletion of antigen-specific T cells as cause for the reduced efficacy, but observed a local expansion of antigen-specific regulatory T cells, which could suppress the activation of effector cells. In conclusion, DNA vaccines encoding DEC205-targeted antigens induce peripheral tolerance rather than immunity in our study. Finally, we evaluated our DNA vaccines as prophylactic or therapeutic treatment in an allergen-induced asthma mouse model., (© 2015 John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
29. Codon-optimized filovirus DNA vaccines delivered by intramuscular electroporation protect cynomolgus macaques from lethal Ebola and Marburg virus challenges.
- Author
-
Grant-Klein RJ, Altamura LA, Badger CV, Bounds CE, Van Deusen NM, Kwilas SA, Vu HA, Warfield KL, Hooper JW, Hannaman D, Dupuy LC, and Schmaljohn CS
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Codon, Disease Models, Animal, Drug Evaluation, Preclinical, Enzyme-Linked Immunosorbent Assay, Enzyme-Linked Immunospot Assay, Female, Filoviridae genetics, Glycoproteins genetics, Glycoproteins immunology, Immunoglobulin G blood, Injections, Intramuscular, Interferon-gamma metabolism, Leukocytes, Mononuclear immunology, Macaca fascicularis, Male, Neutralization Tests, Plasmids, Survival Analysis, Treatment Outcome, Vaccines, DNA administration & dosage, Vaccines, DNA genetics, Electroporation, Filoviridae immunology, Hemorrhagic Fever, Ebola prevention & control, Marburg Virus Disease prevention & control, Vaccines, DNA immunology
- Abstract
Cynomolgus macaques were vaccinated by intramuscular electroporation with DNA plasmids expressing codon-optimized glycoprotein (GP) genes of Ebola virus (EBOV) or Marburg virus (MARV) or a combination of codon-optimized GP DNA vaccines for EBOV, MARV, Sudan virus and Ravn virus. When measured by ELISA, the individual vaccines elicited slightly higher IgG responses to EBOV or MARV than did the combination vaccines. No significant differences in immune responses of macaques given the individual or combination vaccines were measured by pseudovirion neutralization or IFN-γ ELISpot assays. Both the MARV and mixed vaccines were able to protect macaques from lethal MARV challenge (5/6 vs. 6/6). In contrast, a greater proportion of macaques vaccinated with the EBOV vaccine survived lethal EBOV challenge in comparison to those that received the mixed vaccine (5/6 vs. 1/6). EBOV challenge survivors had significantly higher pre-challenge neutralizing antibody titers than those that succumbed.
- Published
- 2015
- Full Text
- View/download PDF
30. Novel vaccine regimen elicits strong airway immune responses and control of respiratory syncytial virus in nonhuman primates.
- Author
-
Grunwald T, Tenbusch M, Schulte R, Raue K, Wolf H, Hannaman D, de Swart RL, Uberla K, and Stahl-Hennig C
- Subjects
- Animals, Antibodies, Viral immunology, Disease Models, Animal, Female, Humans, Immunization, Macaca mulatta, Male, Respiratory Syncytial Virus Infections prevention & control, Respiratory Syncytial Virus Infections virology, Respiratory Syncytial Virus Vaccines administration & dosage, Respiratory Syncytial Virus Vaccines genetics, Respiratory Syncytial Virus, Human genetics, Respiratory System virology, Respiratory Syncytial Virus Infections immunology, Respiratory Syncytial Virus Vaccines immunology, Respiratory Syncytial Virus, Human immunology, Respiratory System immunology
- Abstract
Unlabelled: Induction of long-lasting immunity against viral respiratory tract infections remains an elusive goal. Using a nonhuman primate model of human respiratory syncytial virus (hRSV) infection, we compared mucosal and systemic immune responses induced by different DNA delivery approaches to a novel parenteral DNA prime-tonsillar adenoviral vector booster immunization regimen. Intramuscular (i.m.) electroporation (EP) of a DNA vaccine encoding the fusion protein of hRSV induced stronger systemic immune responses than intradermal EP, tattoo immunization, and conventional i.m. DNA injection. A single EP i.m., followed by two atraumatic tonsillar immunizations with the adenoviral vector, elicited strong systemic immune responses, an unique persistent CD4(+) and CD8(+) T cell response in the lower respiratory tract and protection from intranasal hRSV challenge. Thus, parenteral DNA priming followed by booster immunization targeted to a mucosal inductive site constitutes an effective vaccine regimen for eliciting protective immune responses at mucosal effector sites., Importance: The human respiratory syncytial virus (hRSV) is the most common cause of severe respiratory tract disease in infancy and leads to substantial morbidity and morality in the elderly. In this study, we compared the immunogenicity and efficacy of several gene-based immunization protocols in rhesus macaques. Thereby, we found that the combination of an initially parenterally delivered DNA vaccine with a subsequent atraumatic tonsillar adenoviral vector immunization results in a strong systemic immune response accompanied by an exceptional high T-cell response in the mucosa. Strikingly, these animals were protected against a RSV challenge infection controlling the viral replication indicated by a 1,000-fold-lower viral load in the lower respiratory tract. Since mucosal cellular responses of this strength had not been described in earlier RSV vaccine studies, this heterologous DNA prime-tonsillar boost vaccine strategy is very promising and should be pursued for further preclinical and clinical testing.
- Published
- 2014
- Full Text
- View/download PDF
31. Control of HPV-associated tumors by innovative therapeutic HPV DNA vaccine in the absence of CD4+ T cells.
- Author
-
Peng S, Song L, Knoff J, Wang JW, Chang YN, Hannaman D, Wu TC, Alvarez RD, Roden RB, and Hung CF
- Abstract
Human papillomavirus (HPV) infections are particularly problematic for HIV + and solid organ transplant patients with compromised CD4+ T cell-dependent immunity as they produce more severe and progressive disease compared to healthy individuals. There are no specific treatments for chronic HPV infection, resulting in an urgent unmet need for a modality that is safe and effective for both immunocompromised and otherwise normal patients with recalcitrant disease. DNA vaccination is attractive because it avoids the risks of administration of live vectors to immunocompromised patients, and can induce potent HPV-specific cytotoxic T cell responses. We have developed a DNA vaccine (pNGVL4a-hCRTE6E7L2) encoding calreticulin (CRT) fused to E6, E7 and L2 proteins of HPV-16, the genotype associated with approximately 90% vaginal, vulvar, anal, penile and oropharyngeal HPV-associated cancers and the majority of cervical cancers. Administration of the DNA vaccine by intramuscular (IM) injection followed by electroporation induced significantly greater HPV-specific immune responses compared to IM injection alone or mixed with alum. Furthermore, pNGVL4a-hCRTE6E7L2 DNA vaccination via electroporation of mice carrying an intravaginal HPV-16 E6/E7-expressing syngeneic tumor demonstrated more potent therapeutic effects than IM vaccination alone. Of note, administration of the DNA vaccine by IM injection followed by electroporation elicited potent E6 and E7-specific CD8+ T cell responses and antitumor effects despite CD4+ T cell-depletion, although no antibody response was detected. While CD4+ T cell-depletion did reduce the E6 and E7-specific CD8+ T cell response, it remained sufficient to prevent subcutaneous tumor growth and to eliminate circulating tumor cells in a model of metastatic HPV-16+ cancer. Thus, the antibody response was CD4-dependent, whereas CD4+ T cell help enhanced the E6/E7-specific CD8+ T cell immunity, but was not required. Taken together, our data suggest that pNGVL4a-hCRTE6E7L2 DNA vaccination via electroporation warrants testing in otherwise healthy patients and those with compromised CD4+ T cell immunity to treat HPV-16-associated anogenital disease and cancer.
- Published
- 2014
- Full Text
- View/download PDF
32. Immunologic responses to xenogeneic tyrosinase DNA vaccine administered by electroporation in patients with malignant melanoma.
- Author
-
Yuan J, Ku GY, Adamow M, Mu Z, Tandon S, Hannaman D, Chapman P, Schwartz G, Carvajal R, Panageas KS, Houghton AN, and Wolchok JD
- Abstract
Background: Prior studies show that intramuscular injection and particle-mediated epidermal delivery of xenogeneic melanosomal antigens (tyrosinase or Tyr, gp100) induce CD8(+) T cell responses to the syngeneic protein. To further define the optimal vaccination strategy, we conducted a phase I study of in vivo electroporation (EP) of a murine Tyr DNA vaccine (pINGmuTyr) in malignant melanoma patients., Methods: Human leukocyte antigen (HLA)-A1, A2, A24 or B35 stage IIb-IV melanoma patients received up to five doses of the mouse tyrosinase DNA vaccine by EP every three weeks at dose levels of 0.2 mg, 0.5 mg, or 1.5 mg per injection. Peripheral blood mononuclear cells (PBMC) were collected, cultured with a peptide pool containing eight HLA class I-restricted Tyr-specific T-cell epitopes, and analyzed by HLA-A*0101-restricted tetramers and intracellular cytokine staining (ICS)., Results: Twenty-four patients received ≥1 dose of the pINGmuTyr vaccine; PBMCs from 21 patients who completed all five doses were available for Tyr immune assays. The only common toxicity was grade 1 injection site reaction. Six of 15 patients (40%) in the 1.5 mg dose cohort developed Tyr-reactive CD8(+) T cell responses following stimulation, defined as a ≥3 standard deviation increase in baseline reactivity by tetramer or ICS assays. No Tyr-reactive CD8(+) T cell response was detected in the 0.2 mg and 0.5 mg dose cohort patients. Epitope spreading of CD8(+) T cell response to NY-ESO-1 was observed in one patient with vitiligo. One patient subsequently received ipilimumab and developed an enhanced Tyr-reactive response with polyfunctional cytokine profile. After a median follow-up of 40.9 months, median survival has not been reached., Conclusions: A regimen of five immunizations with pINGmuTyr administered by EP was found to be safe and resulted in Tyr-reactive immune responses in six of 15 patients at 1.5 mg dose cohort., Trial Registration: ClinicalTrials.gov NCT00471133.
- Published
- 2013
- Full Text
- View/download PDF
33. Protective efficacy and immunogenicity of a combinatory DNA vaccine against Influenza A Virus and the Respiratory Syncytial Virus.
- Author
-
Stab V, Nitsche S, Niezold T, Storcksdieck Genannt Bonsmann M, Wiechers A, Tippler B, Hannaman D, Ehrhardt C, Uberla K, Grunwald T, and Tenbusch M
- Subjects
- Animals, Antigens, Viral immunology, Female, Immunity, Cellular immunology, Immunity, Humoral immunology, Immunization, Mice, Mice, Inbred BALB C, Vaccines, Combined immunology, Influenza A virus immunology, Respiratory Syncytial Viruses immunology, Vaccines, DNA immunology
- Abstract
The Respiratory Syncytial Virus (RSV) and Influenza A Virus (IAV) are both two major causative agents of severe respiratory tract infections in humans leading to hospitalization and thousands of deaths each year. In this study, we evaluated the immunogenicity and efficacy of a combinatory DNA vaccine in comparison to the single component vaccines against both diseases in a mouse model. Intramuscular electroporation with plasmids expressing the hemagglutinin (HA) of IAV and the F protein of RSV induced strong humoral immune responses regardless if they were delivered in combination or alone. In consequence, high neutralizing antibody titers were detected, which conferred protection against a lethal challenge with IAV. Furthermore, the viral load in the lungs after a RSV infection could be dramatically reduced in vaccinated mice. Concurrently, substantial amounts of antigen-specific, polyfunctional CD8⁺ T-cells were measured after vaccination. Interestingly, the cellular response to the hemagglutinin was significantly reduced in the presence of the RSV-F encoding plasmid, but not vice versa. Although these results indicate a suppressive effect of the RSV-F protein, the protective efficacy of the combinatory vaccine was comparable to the efficacy of both single-component vaccines. In conclusion, the novel combinatory vaccine against RSV and IAV may have great potential to reduce the rate of severe respiratory tract infections in humans without increasing the number of necessary vaccinations.
- Published
- 2013
- Full Text
- View/download PDF
34. A single electroporation delivery of a DNA vaccine containing the hemagglutinin gene of Asian H5N1 avian influenza virus generated a protective antibody response in chickens against a North American virus strain.
- Author
-
Ogunremi O, Pasick J, Kobinger GP, Hannaman D, Berhane Y, Clavijo A, and van Drunen Littel-van den Hurk S
- Subjects
- Animals, Chickens, Enzyme-Linked Immunosorbent Assay, Hemagglutination Inhibition Tests, Hemagglutinin Glycoproteins, Influenza Virus genetics, Influenza A Virus, H5N1 Subtype genetics, Influenza Vaccines administration & dosage, Influenza Vaccines genetics, Influenza in Birds immunology, Influenza in Birds pathology, Influenza in Birds virology, Survival Analysis, Vaccines, DNA administration & dosage, Vaccines, DNA genetics, Viral Load, Antibodies, Viral blood, Electroporation methods, Hemagglutinin Glycoproteins, Influenza Virus immunology, Influenza A Virus, H5N1 Subtype immunology, Influenza Vaccines immunology, Influenza in Birds prevention & control, Vaccines, DNA immunology
- Abstract
Protection against the avian influenza (AI) H5N1 virus is suspected to be mainly conferred by the presence of antibodies directed against the hemagglutinin (HA) protein of the virus. A single electroporation delivery of 100 or 250 μg of a DNA vaccine construct, pCAG-HA, carrying the HA gene of strain A/Hanoi/30408/2005 (H5N1), in chickens led to the development of anti-HA antibody response in 16 of 17 immunized birds, as measured by a hemagglutination inhibition (HI) test, competitive enzyme-linked immunosorbent assay (cELISA), and an indirect ELISA. Birds vaccinated by electroporation (n = 11) were protected from experimental AI challenge with strain A/chicken/Pennsylvania/1370/1/1983 (H5N2) as judged by low viral load, absence of clinical symptoms, and absence of mortality (n = 11). In contrast, only two out of 10 birds vaccinated with the same vaccine dose (100 or 250 μg) but without electroporation developed antibodies. These birds showed high viral loads and significant morbidity and mortality after the challenge. Seroconversion was reduced in birds electroporated with a low vaccine dose (10 μg), but the antibody-positive birds were protected against virus challenge. Nonelectroporation delivery of a low-dose vaccine did not result in seroconversion, and the birds were as susceptible as those in the control groups that received the control pCAG vector. Electroporation delivery of the DNA vaccine led to enhanced antibody responses and to protection against the AI virus challenge. The HI test, cELISA, or indirect ELISA for anti-H5 antibodies might serve as a good predictor of the potency and efficacy of a DNA immunization strategy against AI in chickens.
- Published
- 2013
- Full Text
- View/download PDF
35. Two doses of bovine viral diarrhea virus DNA vaccine delivered by electroporation induce long-term protective immune responses.
- Author
-
van Drunen Littel-van den Hurk S, Lawman Z, Snider M, Wilson D, van den Hurk JV, Ellefsen B, and Hannaman D
- Subjects
- Animals, Cattle, Cattle Diseases immunology, Cattle Diseases prevention & control, Cattle Diseases virology, Diarrhea Viruses, Bovine Viral genetics, Immunization Schedule, Vaccines, DNA administration & dosage, Vaccines, DNA immunology, Viral Load, Viral Vaccines administration & dosage, Bovine Virus Diarrhea-Mucosal Disease genetics, Bovine Virus Diarrhea-Mucosal Disease immunology, Bovine Virus Diarrhea-Mucosal Disease prevention & control, Diarrhea Viruses, Bovine Viral immunology, Electroporation, Immunization veterinary, Viral Vaccines immunology
- Abstract
Bovine viral diarrhea virus (BVDV) is a pathogen of major importance in cattle, so there is a need for new effective vaccines. DNA vaccines induce balanced immune responses and are relatively inexpensive and thus promising for both human and veterinary applications. In this study, newborn calves with maternal antibodies were vaccinated intramuscularly (i.m.) with a BVDV E2 DNA vaccine with the TriGrid Delivery System for i.m. delivery (TDS-IM). Two doses of this vaccine spaced 6 or 12 weeks apart were sufficient to induce significant virus-neutralizing antibody titers, numbers of activated T cells, and reduction in viral shedding and clinical presentations after BVDV-2 challenge. In contrast to the placebo-treated animals, the vaccinated calves did not lose any weight, which is an excellent indicator of the well-being of an animal and has a significant economic impact. Furthermore, the interval between the two vaccinations did not influence the magnitude of the immune responses or degree of clinical protection, and a third immunization was not necessary or beneficial. Since electroporation may enhance not only the magnitude but also the duration of immunity after DNA immunization, the interval between vaccination and challenge was extended in a second trial, which showed that two doses of this E2 DNA vaccine again significantly reduced clinical disease against BVDV for several months. These results are promising and support this technology for use against infectious diseases in cattle and large species, including humans, in general.
- Published
- 2013
- Full Text
- View/download PDF
36. Low-dose cyclophosphamide administered as daily or single dose enhances the antitumor effects of a therapeutic HPV vaccine.
- Author
-
Peng S, Lyford-Pike S, Akpeng B, Wu A, Hung CF, Hannaman D, Saunders JR, Wu TC, and Pai SI
- Subjects
- Animals, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines administration & dosage, Combined Modality Therapy, Flow Cytometry, Mice, Mice, Inbred C57BL, Neoplasms, Experimental immunology, Papillomavirus Vaccines administration & dosage, Vaccines, DNA administration & dosage, Vaccines, DNA immunology, Antineoplastic Agents administration & dosage, Cancer Vaccines immunology, Cyclophosphamide administration & dosage, Neoplasms, Experimental therapy, Papillomavirus Vaccines immunology
- Abstract
Although therapeutic HPV vaccines are able to elicit systemic HPV-specific immunity, clinical responses have not always correlated with levels of vaccine-induced CD8(+) T cells in human clinical trials. This observed discrepancy may be attributable to an immunosuppressive tumor microenvironment in which the CD8(+) T cells are recruited. Regulatory T cells (Tregs) are cells that can dampen cytotoxic CD8(+) T-cell function. Cyclophosphamide (CTX) is a systemic chemotherapeutic agent, which can eradicate immune cells, including inhibitory Tregs. The optimal dose and schedule of CTX administration in combination with immunotherapy to eliminate the Treg population without adversely affecting vaccine-induced T-cell responses is unknown. Therefore, we investigated various dosing and administration schedules of CTX in combination with a therapeutic HPV vaccine in a preclinical tumor model. HPV tumor-bearing mice received either a single preconditioning dose or a daily dose of CTX in combination with the pNGVL4a-CRT/E7(detox) DNA vaccine. Both single and daily dosing of CTX in combination with vaccine had a synergistic antitumor effect as compared to monotherapy alone. The potent antitumor responses were attributed to the reduction in Treg frequency and increased infiltration of HPV16 E7-specific CD8(+) T cells, which led to higher ratios of CD8(+)/Treg and CD8(+)/CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSCs). There was an observed trend toward decreased vaccine-induced CD8(+) T-cell frequency with daily dosing of CTX. We recommend a single, preconditioning dose of CTX prior to vaccination due to its efficacy, ease of administration, and reduced cumulative adverse effect on vaccine-induced T cells.
- Published
- 2013
- Full Text
- View/download PDF
37. In vivo electroporation improves therapeutic potency of a DNA vaccine targeting hepadnaviral proteins.
- Author
-
Khawaja G, Buronfosse T, Jamard C, Abdul F, Guerret S, Zoulim F, Luxembourg A, Hannaman D, Evans CF, Hartmann D, and Cova L
- Subjects
- Animals, Antibodies, Viral biosynthesis, Antibodies, Viral immunology, Chronic Disease, DNA, Circular genetics, DNA, Circular immunology, Disease Models, Animal, Ducks, Electroporation, Epitopes, Hepadnaviridae Infections immunology, Hepadnaviridae Infections prevention & control, Hepadnaviridae Infections virology, Hepatitis B Vaccines immunology, Hepatitis, Viral, Animal immunology, Hepatitis, Viral, Animal virology, Immune Tolerance, Immunity, Humoral, Interferon-gamma biosynthesis, Interferon-gamma immunology, Plasmids, Vaccines, DNA immunology, Viral Core Proteins genetics, Viral Envelope Proteins genetics, Viremia immunology, Viremia prevention & control, Viremia veterinary, Viremia virology, Hepadnaviridae Infections veterinary, Hepatitis B Vaccines administration & dosage, Hepatitis B Virus, Duck immunology, Hepatitis, Viral, Animal prevention & control, Vaccines, DNA administration & dosage, Viral Core Proteins immunology, Viral Envelope Proteins immunology
- Abstract
This preclinical study investigated the therapeutic efficacy of electroporation (EP)-based delivery of plasmid DNA (pDNA) encoding viral proteins (envelope, core) and IFN-γ in the duck model of chronic hepatitis B virus (DHBV) infection. Importantly, only DNA EP-therapy resulted in a significant decrease in mean viremia titers and in intrahepatic covalently closed circular DNA (cccDNA) levels in chronic DHBV-carrier animals, compared with standard needle pDNA injection (SI). In addition, DNA EP-therapy stimulated in all virus-carriers a humoral response to DHBV preS protein, recognizing a broader range of major antigenic regions, including neutralizing epitopes, compared with SI. DNA EP-therapy led also to significant higher intrahepatic IFN-γ RNA levels in DHBV-carriers compared to other groups, in the absence of adverse effects. We provide the first evidence on DNA EP-therapy benefit in terms of hepadnaviral infection clearance and break of immune tolerance in virus-carriers, supporting its clinical application for chronic hepatitis B., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
38. A multiagent filovirus DNA vaccine delivered by intramuscular electroporation completely protects mice from ebola and Marburg virus challenge.
- Author
-
Grant-Klein RJ, Van Deusen NM, Badger CV, Hannaman D, Dupuy LC, and Schmaljohn CS
- Subjects
- Animals, Ebolavirus immunology, Ebolavirus pathogenicity, Female, Hemorrhagic Fever, Ebola immunology, Marburg Virus Disease immunology, Marburgvirus immunology, Marburgvirus pathogenicity, Mice, Mice, Inbred BALB C, Electroporation methods, Hemorrhagic Fever, Ebola prevention & control, Marburg Virus Disease prevention & control, Muscles metabolism, Vaccines, DNA administration & dosage, Vaccines, DNA therapeutic use
- Abstract
We evaluated the immunogenicity and protective efficacy of DNA vaccines expressing the codon-optimized envelope glycoprotein genes of Zaire ebolavirus, Sudan ebolavirus, and Marburg marburgvirus (Musoke and Ravn). Intramuscular or intradermal delivery of the vaccines in BALB/c mice was performed using the TriGrid™ electroporation device. Mice that received DNA vaccines against the individual viruses developed robust glycoprotein-specific antibody titers as determined by ELISA and survived lethal viral challenge with no display of clinical signs of infection. Survival curve analysis revealed there was a statistically significant increase in survival compared to the control groups for both the Ebola and Ravn virus challenges. These data suggest that further analysis of the immune responses generated in the mice and additional protection studies in nonhuman primates are warranted.
- Published
- 2012
- Full Text
- View/download PDF
39. A DNA-based candidate HIV vaccine delivered via in vivo electroporation induces CD4 responses toward the α4β7-binding V2 loop of HIV gp120 in healthy volunteers.
- Author
-
Kopycinski J, Cheeseman H, Ashraf A, Gill D, Hayes P, Hannaman D, Gilmour J, Cox JH, and Vasan S
- Subjects
- Adult, HIV Envelope Protein gp120 genetics, HIV-1 genetics, Healthy Volunteers, Humans, Inulin administration & dosage, Inulin adverse effects, Inulin immunology, Vaccination methods, Vaccines, DNA administration & dosage, Vaccines, DNA adverse effects, CD4-Positive T-Lymphocytes immunology, Electroporation methods, HIV Envelope Protein gp120 immunology, HIV-1 immunology, Inulin analogs & derivatives, Vaccines, DNA immunology
- Abstract
Administration of a clade C/B' candidate HIV-1 DNA vaccine, ADVAX, by in vivo electroporation (EP) was safe and more immunogenic than intramuscular administration without EP. The breadth and specificity of T-cell responses to full-length Env were mapped. Responses to multiple Env regions were induced, with most focusing on V3/C4 and V2 regions, including the α4β7 integrin-binding domain. The breadth of responses induced by this DNA vaccine regimen was comparable to that of viral-vectored vaccine regimens.
- Published
- 2012
- Full Text
- View/download PDF
40. Enhanced magnitude and breadth of neutralizing humoral response to a DNA vaccine targeting the DHBV envelope protein delivered by in vivo electroporation.
- Author
-
Khawaja G, Buronfosse T, Jamard C, Guerret S, Zoulim F, Luxembourg A, Hannaman D, Evans C, Hartmann D, and Cova L
- Subjects
- Amino Acid Sequence, Animals, Drug Delivery Systems, Ducks, Epitopes, B-Lymphocyte immunology, Hepadnaviridae Infections prevention & control, Hepadnaviridae Infections virology, Hepatitis B Antibodies blood, Hepatitis B Virus, Duck genetics, Hepatitis, Viral, Animal prevention & control, Hepatitis, Viral, Animal virology, Humans, Molecular Sequence Data, Neutralization Tests, Protein Precursors chemistry, Protein Precursors immunology, Vaccines, DNA immunology, Antibodies, Neutralizing blood, Electroporation methods, Hepadnaviridae Infections immunology, Hepatitis B Virus, Duck immunology, Hepatitis, Viral, Animal immunology, Vaccines, DNA administration & dosage, Viral Envelope Proteins immunology
- Abstract
We explored in the duck hepatitis B virus (DHBV) model the impact of electroporation (EP)-mediated DNA vaccine delivery on the neutralizing humoral response to viral preS/S large envelope protein. EP enhanced the kinetics and magnitude of anti-preS response compared to the standard needle DNA injection (SI). Importantly, EP dramatically enhanced the neutralizing potency of the humoral response, since antibodies induced by low DNA dose (10 μg) were able to highly neutralize DHBV and to recognize ten antigenic regions, including four neutralization epitopes. Whereas, SI-induced antibodies by the same low DNA dose were not neutralizing and the epitope pattern was extremely narrow, since it was limited to only one epitope. Thus, EP-based delivery was able to improve the dose efficiency of DNA vaccine and to maintain a highly neutralizing, multi-specific B-cell response, suggesting that it may be an effective approach for chronic hepatitis B therapy at clinically feasible DNA dose., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
41. Immunogenicity of DNA vaccines encoding simian immunodeficiency virus antigen targeted to dendritic cells in rhesus macaques.
- Author
-
Tenbusch M, Ignatius R, Nchinda G, Trumpfheller C, Salazar AM, Töpfer K, Sauermann U, Wagner R, Hannaman D, Tenner-Racz K, Racz P, Stahl-Hennig C, and Uberla K
- Subjects
- Animals, Immunity, Cellular, Macaca mulatta, Antigens, Viral immunology, Dendritic Cells immunology, Simian Immunodeficiency Virus immunology, Vaccines, DNA immunology
- Abstract
Background: Targeting antigens encoded by DNA vaccines to dendritic cells (DCs) in the presence of adjuvants enhances their immunogenicity and efficacy in mice., Methodology/principal Findings: To explore the immunogenicity of this approach in non-human primates, we generated a single chain antibody to the antigen uptake receptor DEC-205 expressed on rhesus macaque DCs. DNA vaccines encoding this single chain antibody fused to the SIV capsid protein were delivered to six monkeys each by either intramuscular electroporation or conventional intramuscular injection co-injected or not with poly ICLC, a stabilized poly I: C analogue, as adjuvant. Antibodies to capsid were induced by the DC-targeting and non-targeting control DNA delivered by electroporation while conventional DNA immunization at a 10-fold higher dose of DNA failed to induce detectable humoral immune responses. Substantial cellular immune responses were also observed after DNA electroporation of both DNAs, but stronger responses were induced by the non-targeting vaccine. Conventional immunization with the DC-targeting DNA at a 10-fold higher dose did not give rise to substantial cellular immune responses, neither when co-injected with poly ICLC., Conclusions/significance: The study confirms the potent immunogenicity of DNA vaccines delivered by electroporation. Targeting the DNA via a single chain antibody to DEC-205 expressed by DCs, however, does not improve the immunogenicity of the antigens in non-human primates.
- Published
- 2012
- Full Text
- View/download PDF
42. A DNA vaccine for venezuelan equine encephalitis virus delivered by intramuscular electroporation elicits high levels of neutralizing antibodies in multiple animal models and provides protective immunity to mice and nonhuman primates.
- Author
-
Dupuy LC, Richards MJ, Ellefsen B, Chau L, Luxembourg A, Hannaman D, Livingston BD, and Schmaljohn CS
- Subjects
- Animals, Disease Models, Animal, Electroporation, Encephalitis Virus, Venezuelan Equine genetics, Encephalomyelitis, Venezuelan Equine pathology, Female, Fever prevention & control, Glycoproteins genetics, Glycoproteins immunology, Lymphopenia prevention & control, Macaca, Male, Mice, Mice, Inbred BALB C, Rabbits, Recombinant Proteins genetics, Recombinant Proteins immunology, Time Factors, Vaccines, DNA administration & dosage, Vaccines, DNA genetics, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Viral Vaccines administration & dosage, Viral Vaccines genetics, Viremia prevention & control, Antibodies, Neutralizing blood, Antibodies, Viral blood, Encephalitis Virus, Venezuelan Equine immunology, Encephalomyelitis, Venezuelan Equine prevention & control, Vaccines, DNA immunology, Viral Vaccines immunology
- Abstract
We evaluated the immunogenicity and protective efficacy of a DNA vaccine expressing codon-optimized envelope glycoprotein genes of Venezuelan equine encephalitis virus (VEEV) when delivered by intramuscular electroporation. Mice vaccinated with the DNA vaccine developed robust VEEV-neutralizing antibody responses that were comparable to those observed after administration of the live-attenuated VEEV vaccine TC-83 and were completely protected from a lethal aerosol VEEV challenge. The DNA vaccine also elicited strong neutralizing antibody responses in rabbits that persisted at high levels for at least 6 months and could be boosted by a single additional electroporation administration of the DNA performed approximately 6 months after the initial vaccinations. Cynomolgus macaques that received the vaccine by intramuscular electroporation developed substantial neutralizing antibody responses and after an aerosol challenge had no detectable serum viremia and had reduced febrile reactions, lymphopenia, and clinical signs of disease compared to those of negative-control macaques. Taken together, our results demonstrate that this DNA vaccine provides a potent means of protecting against VEEV infections and represents an attractive candidate for further development.
- Published
- 2011
- Full Text
- View/download PDF
43. In vivo electroporation enhances the immunogenicity of an HIV-1 DNA vaccine candidate in healthy volunteers.
- Author
-
Vasan S, Hurley A, Schlesinger SJ, Hannaman D, Gardiner DF, Dugin DP, Boente-Carrera M, Vittorino R, Caskey M, Andersen J, Huang Y, Cox JH, Tarragona-Fiol T, Gill DK, Cheeseman H, Clark L, Dally L, Smith C, Schmidt C, Park HH, Kopycinski JT, Gilmour J, Fast P, Bernard R, and Ho DD
- Subjects
- AIDS Vaccines pharmacology, Adolescent, Adult, Cytokines metabolism, Double-Blind Method, Electroporation standards, Female, Humans, Injections, Intramuscular, Male, Middle Aged, T-Lymphocytes immunology, Vaccines, DNA pharmacology, Young Adult, AIDS Vaccines administration & dosage, Electroporation methods, HIV-1 immunology, Immunity, Cellular drug effects, Vaccines, DNA administration & dosage
- Abstract
Background: DNA-based vaccines have been safe but weakly immunogenic in humans to date., Methods and Findings: We sought to determine the safety, tolerability, and immunogenicity of ADVAX, a multigenic HIV-1 DNA vaccine candidate, injected intramuscularly by in vivo electroporation (EP) in a Phase-1, double-blind, randomized placebo-controlled trial in healthy volunteers. Eight volunteers each received 0.2 mg, 1 mg, or 4 mg ADVAX or saline placebo via EP, or 4 mg ADVAX via standard intramuscular injection at weeks 0 and 8. A third vaccination was administered to eleven volunteers at week 36. EP was safe, well-tolerated and considered acceptable for a prophylactic vaccine. EP delivery of ADVAX increased the magnitude of HIV-1-specific cell mediated immunity by up to 70-fold over IM injection, as measured by gamma interferon ELISpot. The number of antigens to which the response was detected improved with EP and increasing dosage. Intracellular cytokine staining analysis of ELISpot responders revealed both CD4+ and CD8+ T cell responses, with co-secretion of multiple cytokines., Conclusions: This is the first demonstration in healthy volunteers that EP is safe, tolerable, and effective in improving the magnitude, breadth and durability of cellular immune responses to a DNA vaccine candidate., Trial Registration: ClinicalTrials.gov NCT00545987.
- Published
- 2011
- Full Text
- View/download PDF
44. The efficacy of DNA vaccination is enhanced in mice by targeting the encoded protein to dendritic cells.
- Author
-
Nchinda G, Kuroiwa J, Oks M, Trumpfheller C, Park CG, Huang Y, Hannaman D, Schlesinger SJ, Mizenina O, Nussenzweig MC, Uberla K, and Steinman RM
- Subjects
- Animals, Antibodies immunology, Antigens genetics, Cell Line, Cricetinae, Gene Products, gag genetics, Gene Products, gag immunology, Gene Products, gag metabolism, Humans, Mice, Mucous Membrane immunology, T-Lymphocytes immunology, Antigens immunology, Antigens metabolism, Dendritic Cells immunology, Vaccines, DNA immunology
- Abstract
DNA vaccines promote an immune response by providing antigen-encoding DNA to the recipient, but the efficacy of such vaccines needs improving. Many approaches have considerable potential but currently induce relatively weak immune responses despite multiple high doses of DNA vaccine. Here, we asked whether targeting vaccine antigens to DCs would increase the immunity and protection that result from DNA vaccines. To determine this, we generated a DNA vaccine encoding a fusion protein comprised of the vaccine antigen and a single-chain Fv antibody (scFv) specific for the DC-restricted antigen-uptake receptor DEC205. Following vaccination of mice, the vaccine antigen was expressed selectively by DCs, which were required for the increased efficacy of MHC class I and MHC class II antigen presentation relative to a control scFv DNA vaccine. In addition, a DNA vaccine encoding an HIV gag p41-scFv DEC205 fusion protein induced 10-fold higher antibody levels and increased numbers of IFN-gamma-producing CD4+ and CD8+ T cells. After a single i.m. injection of the DNA vaccine encoding an HIV gag p41-scFv DEC205 fusion protein, mice were protected from an airway challenge with a recombinant vaccinia virus expressing the HIV gag p41, even with 1% of the dose of nontargeted DNA vaccine. The efficacy of DNA vaccines therefore may be enhanced by inclusion of sequences such as single-chain antibodies to target the antigen to DCs.
- Published
- 2008
- Full Text
- View/download PDF
45. Gene-based intramuscular interferon-beta therapy for experimental autoimmune encephalomyelitis.
- Author
-
Jaini R, Hannaman D, Johnson JM, Bernard RM, Altuntas CZ, Delasalas MM, Kesaraju P, Luxembourg A, Evans CF, and Tuohy VK
- Subjects
- 2',5'-Oligoadenylate Synthetase analysis, 2',5'-Oligoadenylate Synthetase genetics, 2',5'-Oligoadenylate Synthetase metabolism, Animals, Electroporation, Female, Gene Transfer Techniques, Interferon Type I administration & dosage, Mice, Mice, Inbred Strains, Muscle, Skeletal, Recombinant Proteins, Spleen enzymology, Encephalomyelitis, Autoimmune, Experimental therapy, Genetic Therapy, Interferon-beta genetics, Multiple Sclerosis therapy
- Abstract
In contrast to serial injections of recombinant interferon-beta (IFN-beta) for long-term therapy of multiple sclerosis (MS), prolonged systemic delivery of proteins derived through in vivo gene transfer may provide a more clinically relevant alternative. Here we compare the therapeutic efficacies of electroporation (EP)-mediated intramuscular IFN-beta gene transfer with repeated alternate-day injections of recombinant IFN-beta after the onset of relapsing-remitting experimental autoimmune encephalomyelitis (EAE), an animal model widely used in MS research. We show for the first time that a single EP-mediated intramuscular administration of 20 microg of an IFN-beta-expressing plasmid provides long-term expression of interferon-inducible genes and is therapeutic in ongoing established EAE. The achieved therapeutic effects of IFN-beta gene delivery were comparable to an 8-week regimen of 10,000 IU rIFN-beta injected every other day and involved a significant inhibition of disease progression and a significant reduction of EAE relapses compared to untreated or null-vector-treated mice. Our results indicate the viability of a convenient and effective gene-based alternative for long-term IFN-beta protein therapy in MS.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.