1
|
Mattathil JG, Volz A, Onabajo OO, Maynard S, Bixler SL, Shen XX, Vargas-Inchaustegui D, Robert-Guroff M, Lebranche C, Tomaras G, Montefiori D, Sutter G, Mattapallil JJ. Direct intranodal tonsil vaccination with modified vaccinia Ankara vaccine protects macaques from highly pathogenic SIVmac251. Nat Commun 2023; 14:1264. [PMID: 36882405 PMCID: PMC9990026 DOI: 10.1038/s41467-023-36907-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Human immunodeficiency virus (HIV) is a mucosally transmitted virus that causes immunodeficiency and AIDS. Developing efficacious vaccines to prevent infection is essential to control the epidemic. Protecting the vaginal and rectal mucosa, the primary routes of HIV entry has been a challenge given the significant compartmentalization between the mucosal and peripheral immune systems. We hypothesized that direct intranodal vaccination of mucosa associated lymphoid tissue (MALT) such as the readily accessible palatine tonsils could overcome this compartmentalization. Here we show that rhesus macaques primed with plasmid DNA encoding SIVmac251-env and gag genes followed by an intranodal tonsil MALT boost with MVA encoding the same genes protects from a repeated low dose intrarectal challenge with highly pathogenic SIVmac251; 43% (3/7) of vaccinated macaques remained uninfected after 9 challenges as compared to the unvaccinated control (0/6) animals. One vaccinated animal remained free of infection even after 22 challenges. Vaccination was associated with a ~2 log decrease in acute viremia that inversely correlated with anamnestic immune responses. Our results suggest that a combination of systemic and intranodal tonsil MALT vaccination could induce robust adaptive and innate immune responses leading to protection from mucosal infection with highly pathogenic HIV and rapidly control viral breakthroughs.
Collapse
Affiliation(s)
- Jeffy G Mattathil
- Henry M. Jackson Foundation for Military Medicine, Bethesda, MD, USA
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
| | | | - Sean Maynard
- Henry M. Jackson Foundation for Military Medicine, Bethesda, MD, USA
| | - Sandra L Bixler
- Henry M. Jackson Foundation for Military Medicine, Bethesda, MD, USA
| | | | | | | | | | | | | | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, LMU, Munich, Germany
| | - Joseph J Mattapallil
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA.
| |
Collapse
|
2
|
Araújo NM, Rubio IGS, Toneto NPA, Morale MG, Tamura RE. The use of adenoviral vectors in gene therapy and vaccine approaches. Genet Mol Biol 2022; 45:e20220079. [PMID: 36206378 PMCID: PMC9543183 DOI: 10.1590/1678-4685-gmb-2022-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022] Open
Abstract
Adenovirus was first identified in the 1950s and since then this pathogenic group
of viruses has been explored and transformed into a genetic transfer vehicle.
Modification or deletion of few genes are necessary to transform it into a
conditionally or non-replicative vector, creating a versatile tool capable of
transducing different tissues and inducing high levels of transgene expression.
In the early years of vector development, the application in monogenic diseases
faced several hurdles, including short-term gene expression and even a fatality.
On the other hand, an adenoviral delivery strategy for treatment of cancer was
the first approved gene therapy product. There is an increasing interest in
expressing transgenes with therapeutic potential targeting the cancer hallmarks,
inhibiting metastasis, inducing cancer cell death or modulating the immune
system to attack the tumor cells. Replicative adenovirus as vaccines may be even
older and date to a few years of its discovery, application of non-replicative
adenovirus for vaccination against different microorganisms has been
investigated, but only recently, it demonstrated its full potential being one of
the leading vaccination tools for COVID-19. This is not a new vector nor a new
technology, but the result of decades of careful and intense work in this
field.
Collapse
Affiliation(s)
- Natália Meneses Araújo
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil.
| | - Ileana Gabriela Sanchez Rubio
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | | | - Mirian Galliote Morale
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | - Rodrigo Esaki Tamura
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil.
| |
Collapse
|
3
|
Harris JE. The repeated setbacks of HIV vaccine development laid the groundwork for SARS-CoV-2 vaccines. HEALTH POLICY AND TECHNOLOGY 2022; 11:100619. [PMID: 35340773 PMCID: PMC8935961 DOI: 10.1016/j.hlpt.2022.100619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The decades-long effort to produce a workable HIV vaccine has hardly been a waste of public and private resources. To the contrary, the scientific know-how acquired along the way has served as the critical foundation for the development of vaccines against the novel, pandemic SARS-CoV-2 virus. We retell the real-world story of HIV vaccine research - with all its false leads and missteps - in a way that sheds light on the current state of the art of antiviral vaccines. We find that HIV-related R&D had more than a general spillover effect. In fact, the repeated failures of phase 2 and 3 clinical trials of HIV vaccine candidates have served as a critical stimulus to the development of successful vaccine technologies today. We rebut the counterargument that HIV vaccine development has been no more than a blind alley, and that recently developed vaccines against COVID-19 are really descendants of successful vaccines against Ebola, MERS, and SARS. These successful vaccines likewise owe much to the vicissitudes of HIV vaccine development. We then discuss how the failures of HIV vaccine development have taught us how adapt SARS-CoV-2 vaccines to immune escape from emerging variants. Finally, we inquire whether recent advances in the development of vaccines against SARS-CoV-2 might in turn further the development of an HIV vaccine - what we describe as a reverse spillover effect.
Collapse
Affiliation(s)
- Jeffrey E Harris
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Eisner Health, Los Angeles, CA 90015, USA
| |
Collapse
|
4
|
Wilmschen S, Schmitz JE, Kimpel J. Viral Vectors for the Induction of Broadly Neutralizing Antibodies against HIV. Vaccines (Basel) 2019; 7:vaccines7030119. [PMID: 31546894 PMCID: PMC6789710 DOI: 10.3390/vaccines7030119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 01/10/2023] Open
Abstract
Extensive research on generating an efficient HIV vaccine is ongoing. A major aim of HIV vaccines is the induction of long-lasting, broadly neutralizing antibodies (bnAbs) that can confer sterile immunity for a prolonged period of time. Several strategies have been explored to reach this goal, i.e. protein immunization, DNA, or viral vectors, or a combination thereof. In this review, we give an overview of approaches using viral vectors for the induction of HIV-specific bnAbs. Many pre-clinical studies were performed using various replication-competent and -incompetent vectors. Amongst them, poxviral and adenoviral vectors were the most prevalent ones. In many studies, viral vectors were combined with a DNA prime or a protein boost. However, neutralizing antibodies were mainly induced against the homologous HIV-1 vaccine strain or tier 1 viruses, and in rare cases, against tier 2 viruses, indicating the need for improved antigens and vaccination strategies. Furthermore, we also review next generation Env antigens that are currently being used in protein vaccination approaches and point out how they could be utilized in viral vectors.
Collapse
Affiliation(s)
- Sarah Wilmschen
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
5
|
Chapman R, Jongwe TI, Douglass N, Chege G, Williamson AL. Heterologous prime-boost vaccination with DNA and MVA vaccines, expressing HIV-1 subtype C mosaic Gag virus-like particles, is highly immunogenic in mice. PLoS One 2017; 12:e0173352. [PMID: 28278263 PMCID: PMC5344398 DOI: 10.1371/journal.pone.0173352] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/19/2017] [Indexed: 12/31/2022] Open
Abstract
In an effort to make affordable vaccines suitable for the regions most affected by HIV-1, we have constructed stable vaccines that express an HIV-1 subtype C mosaic Gag immunogen (BCG-GagM, MVA-GagM and DNA-GagM). Mosaic immunogens have been designed to address the tremendous diversity of this virus. Here we have shown that GagM buds from cells infected and transfected with MVA-GagM and DNA-GagM respectively and forms virus-like particles. Previously we showed that a BCG-GagM prime MVA-GagM boost generated strong cellular immune responses in mice. In this study immune responses to the DNA-GagM and MVA-GagM vaccines were evaluated in homologous and heterologous prime-boost vaccinations. The DNA homologous prime boost vaccination elicited predominantly CD8+ T cells while the homologous MVA vaccination induced predominantly CD4+ T cells. A heterologous DNA-GagM prime MVA-GagM boost induced strong, more balanced Gag CD8+ and CD4+ T cell responses and that were predominantly of an effector memory phenotype. The immunogenicity of the mosaic Gag (GagM) was compared to a naturally occurring subtype C Gag (GagN) using a DNA homologous vaccination regimen. DNA-GagN expresses a natural Gag with a sequence that was closest to the consensus sequence of subtype C viruses sampled in South Africa. DNA-GagM homologous vaccination induced cumulative HIV-1 Gag-specific IFN-γ ELISPOT responses that were 6.5-fold higher than those induced by the DNA-GagN vaccination. Similarly, DNA-GagM vaccination generated 7-fold higher levels of cytokine-positive CD8+ T cells than DNA-GagN, indicating that this subtype C mosaic Gag elicits far more potent immune responses than a consensus-type Gag. Cells transfected and infected with DNA-GagM and MVA-GagM respectively, expressed high levels of GagM and produced budding virus-like particles. Our data indicates that a heterologous prime boost regimen using DNA and MVA vaccines expressing HIV-1 subtype C mosaic Gag is highly immunogenic in mice and warrants further investigation in non-human primates.
Collapse
Affiliation(s)
- Ros Chapman
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Tsungai Ivai Jongwe
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicola Douglass
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gerald Chege
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
6
|
Potential To Streamline Heterologous DNA Prime and NYVAC/Protein Boost HIV Vaccine Regimens in Rhesus Macaques by Employing Improved Antigens. J Virol 2016; 90:4133-4149. [PMID: 26865719 DOI: 10.1128/jvi.03135-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/02/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED In a follow-up to the modest efficacy observed in the RV144 trial, researchers in the HIV vaccine field seek to substantiate and extend the results by evaluating other poxvirus vectors and combinations with DNA and protein vaccines. Earlier clinical trials (EuroVacc trials 01 to 03) evaluated the immunogenicity of HIV-1 clade C GagPolNef and gp120 antigens delivered via the poxviral vector NYVAC. These showed that a vaccination regimen including DNA-C priming prior to a NYVAC-C boost considerably enhanced vaccine-elicited immune responses compared to those with NYVAC-C alone. Moreover, responses were improved by using three as opposed to two DNA-C primes. In the present study, we assessed in nonhuman primates whether such vaccination regimens can be streamlined further by using fewer and accelerated immunizations and employing a novel generation of improved DNA-C and NYVAC-C vaccine candidates designed for higher expression levels and more balanced immune responses. Three different DNA-C prime/NYVAC-C+ protein boost vaccination regimens were tested in rhesus macaques. All regimens elicited vigorous and well-balanced CD8(+)and CD4(+)T cell responses that were broad and polyfunctional. Very high IgG binding titers, substantial antibody-dependent cellular cytotoxicity (ADCC), and modest antibody-dependent cell-mediated virus inhibition (ADCVI), but very low neutralization activity, were measured after the final immunizations. Overall, immune responses elicited in all three groups were very similar and of greater magnitude, breadth, and quality than those of earlier EuroVacc vaccines. In conclusion, these findings indicate that vaccination schemes can be simplified by using improved antigens and regimens. This may offer a more practical and affordable means to elicit potentially protective immune responses upon vaccination, especially in resource-constrained settings. IMPORTANCE Within the EuroVacc clinical trials, we previously assessed the immunogenicity of HIV clade C antigens delivered in a DNA prime/NYVAC boost regimen. The trials showed that the DNA prime crucially improved the responses, and three DNA primes with a NYVAC boost appeared to be optimal. Nevertheless, T cell responses were primarily directed toward Env, and humoral responses were modest. The aim of this study was to assess improved antigens for the capacity to elicit more potent and balanced responses in rhesus macaques, even with various simpler immunization regimens. Our results showed that the novel antigens in fact elicited larger numbers of T cells with a polyfunctional profile and a good Env-GagPolNef balance, as well as high-titer and Fc-functional antibody responses. Finally, comparison of the different schedules indicates that a simpler regimen of only two DNA primes and one NYVAC boost in combination with protein may be very efficient, thus showing that the novel antigens allow for easier immunization protocols.
Collapse
|
7
|
An overview of live attenuated recombinant pseudorabies viruses for use as novel vaccines. J Immunol Res 2014; 2014:824630. [PMID: 24995348 PMCID: PMC4068083 DOI: 10.1155/2014/824630] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/23/2014] [Indexed: 11/18/2022] Open
Abstract
Pseudorabies virus (PRV) is a double-stranded, DNA-based swine virus with a genome approximating 150 kb in size. PRV has many nonessential genes which can be replaced with genes encoding heterologous antigens but without deleterious effects on virus propagation. Recombinant PRVs expressing both native and foreign antigens are able to stimulate immune responses. In this paper, we review the current status of live attenuated recombinant PRVs and live PRV-based vector vaccines with potential for controlling viral infections in animals.
Collapse
|
8
|
de Cassan SC, Draper SJ. Recent advances in antibody-inducing poxviral and adenoviral vectored vaccine delivery platforms for difficult disease targets. Expert Rev Vaccines 2014; 12:365-78. [DOI: 10.1586/erv.13.11] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
9
|
Flingai S, Czerwonko M, Goodman J, Kudchodkar SB, Muthumani K, Weiner DB. Synthetic DNA vaccines: improved vaccine potency by electroporation and co-delivered genetic adjuvants. Front Immunol 2013; 4:354. [PMID: 24204366 PMCID: PMC3816528 DOI: 10.3389/fimmu.2013.00354] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/20/2013] [Indexed: 01/07/2023] Open
Abstract
In recent years, DNA vaccines have undergone a number of technological advancements that have incited renewed interest and heightened promise in the field. Two such improvements are the use of genetically engineered cytokine adjuvants and plasmid delivery via in vivo electroporation (EP), the latter of which has been shown to increase antigen delivery by nearly 1000-fold compared to naked DNA plasmid delivery alone. Both strategies, either separately or in combination, have been shown to augment cellular and humoral immune responses in not only mice, but also in large animal models. These promising results, coupled with recent clinical trials that have shown enhanced immune responses in humans, highlight the bright prospects for DNA vaccines to address many human diseases.
Collapse
Affiliation(s)
- Seleeke Flingai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania , Philadelphia, PA , USA
| | | | | | | | | | | |
Collapse
|
10
|
Immunogenicity analysis following human immunodeficiency virus recombinant DNA and recombinant vaccinia virus Tian Tan prime-boost immunization. SCIENCE CHINA-LIFE SCIENCES 2013; 56:531-40. [PMID: 23645103 DOI: 10.1007/s11427-013-4484-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/13/2013] [Indexed: 10/26/2022]
|
11
|
Abstract
Most neutralizing antibodies act at the earliest steps of viral infection and block interaction of the virus with cellular receptors to prevent entry into host cells. The inability to induce neutralizing antibodies to HIV has been a major obstacle to HIV vaccine research since the early days of the epidemic. However, in the past three years, the definition of a neutralizing antibody against HIV has been revolutionized by the isolation of extremely broad and potent neutralizing antibodies from HIV-infected individuals. Considerable hurdles remain for inducing neutralizing antibodies to a protective level after immunization. Meanwhile, novel technologies to bypass the induction of antibodies are being explored to provide prophylactic antibody-based interventions. This review addresses the challenge of inducing HIV neutralizing antibodies upon immunization and considers notable recent advances in the field. A greater understanding of the successes and failures for inducing a neutralizing response upon immunization is required to accelerate the development of an effective HIV vaccine.
Collapse
Affiliation(s)
- Laura E McCoy
- Wohl Virion Centre, Division of Infection and Immunity, University College London, London WC1E 6BT, England, UK
| | | |
Collapse
|
12
|
Affiliation(s)
- Gary J Nabel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
13
|
|
14
|
Abstract
There is renewed optimism that the goal of developing a highly effective AIDS vaccine is attainable. The HIV-1 vaccine field has seen its first trial of a vaccine candidate that prevents infection. Although modest in efficacy, this finding, along with the recent discovery that the human immune system can produce broadly neutralizing antibodies capable of inhibiting greater than 90% of circulating viruses, provides a guide for the rational design of vaccines and protection by passive immunization. Together, these findings will help shape the next generation of HIV vaccines.
Collapse
|
15
|
Hutnick NA, Myles DJF, Hirao L, Scott VL, Ferraro B, Khan AS, Lewis MG, Miller CJ, Bett AJ, Casimiro D, Sardesai NY, Kim JJ, Shiver J, Weiner DB. An optimized SIV DNA vaccine can serve as a boost for Ad5 and provide partial protection from a high-dose SIVmac251 challenge. Vaccine 2012; 30:3202-8. [PMID: 22406458 DOI: 10.1016/j.vaccine.2012.02.069] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 02/21/2012] [Accepted: 02/25/2012] [Indexed: 11/29/2022]
Abstract
One limitation in the development of an improved cellular response needed for an effective HIV-vaccine is the inability to induce robust effector T-cells capable of suppressing a heterologous challenge. To improve cellular immune responses, we examined the ability of an optimized DNA vaccine to boost the cellular immune responses induced by a highly immunogenic Ad5 prime. Five Chinese rhesus macaques received pVax encoding consensus (con) gag/pol/env intramuscularly (IM) with electroporation followed by the Merck Ad5 gag/pol/nef vaccine. A second group of five animals were vaccinated with Merck Ad5 gag/pol/nef followed by pVax gag/pol/env. One year following vaccination, Ad5-prime DNA-boosted monkeys and four unvaccinated controls received an intrarectal challenge with 1000 ID50 SIV(mac)251. The quality and magnitude of the T-cell response was analyzed by ELISpot and polyfunctional flow cytometry. We observed that an Ad5-prime DNA-boost resulted in significantly elevated SIV-specific T-cell responses even compared with animals receiving a DNA-prime Ad5-boost. Ad5 prime DNA boosted animals were capable of suppressing a pathogenic SIV(mac)251 challenge. Peak control correlated with the expansion of HLA-DR(+) CD8(+) T-cells two weeks post-infection. These data illustrate that high optimization of a DNA vaccine can drive of immune responses primed by a robust vector system. This previously unachievable feature of these newly optimized DNAs warrants future studies of this strategy that may circumvent issues of serology associated with viral vector prime-boost systems.
Collapse
Affiliation(s)
- Natalie A Hutnick
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, School of Medicine, Philadelphia, PA, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Genmei L, Manlin L, Ruiai C, Hongliang H, Dangshuai P. Construction and immunogenicity of recombinant adenovirus expressing ORF2 of PCV2 and porcine IFN gamma. Vaccine 2011; 29:8677-82. [DOI: 10.1016/j.vaccine.2011.08.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 08/17/2011] [Accepted: 08/28/2011] [Indexed: 10/17/2022]
|
17
|
Girard MP, Osmanov S, Assossou OM, Kieny MP. Human immunodeficiency virus (HIV) immunopathogenesis and vaccine development: a review. Vaccine 2011; 29:6191-218. [PMID: 21718747 DOI: 10.1016/j.vaccine.2011.06.085] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/20/2011] [Accepted: 06/22/2011] [Indexed: 02/08/2023]
Abstract
The development of a safe, effective and globally affordable HIV vaccine offers the best hope for the future control of the HIV-1 pandemic. Since 1987, scores of candidate HIV-1 vaccines have been developed which elicited varying degrees of protective responses in nonhuman primate models, including DNA vaccines, subunit vaccines, live vectored recombinant vaccines and various prime-boost combinations. Four of these candidate vaccines have been tested for efficacy in human volunteers, but, to the exception of the recent RV144 Phase III trial in Thailand, which elicited a modest but statistically significant level of protection against infection, none has shown efficacy in preventing HIV-1 infection or in controlling virus replication and delaying progression of disease in humans. Protection against infection was observed in the RV144 trial, but intensive research is needed to try to understand the protective immune mechanisms at stake. Building-up on the results of the RV144 trial and deciphering what possibly are the immune correlates of protection are the top research priorities of the moment, which will certainly accelerate the development of an highly effective vaccine that could be used in conjunction with other HIV prevention and treatment strategies. This article reviews the state of the art of HIV vaccine development and discusses the formidable scientific challenges met in this endeavor, in the context of a better understanding of the immunopathogenesis of the disease.
Collapse
Affiliation(s)
- Marc P Girard
- University Paris 7, French National Academy of Medicine, 39 rue Seignemartin, FR 69008 Lyon, France.
| | | | | | | |
Collapse
|
18
|
Wilks AB, Christian EC, Seaman MS, Sircar P, Carville A, Gomez CE, Esteban M, Pantaleo G, Barouch DH, Letvin NL, Permar SR. Robust vaccine-elicited cellular immune responses in breast milk following systemic simian immunodeficiency virus DNA prime and live virus vector boost vaccination of lactating rhesus monkeys. THE JOURNAL OF IMMUNOLOGY 2010; 185:7097-106. [PMID: 21041730 DOI: 10.4049/jimmunol.1002751] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Breast milk transmission of HIV remains an important mode of infant HIV acquisition. Enhancement of mucosal HIV-specific immune responses in milk of HIV-infected mothers through vaccination may reduce milk virus load or protect against virus transmission in the infant gastrointestinal tract. However, the ability of HIV/SIV strategies to induce virus-specific immune responses in milk has not been studied. In this study, five uninfected, hormone-induced lactating, Mamu A*01(+) female rhesus monkey were systemically primed and boosted with rDNA and the attenuated poxvirus vector, NYVAC, containing the SIVmac239 gag-pol and envelope genes. The monkeys were boosted a second time with a recombinant Adenovirus serotype 5 vector containing matching immunogens. The vaccine-elicited immunodominant epitope-specific CD8(+) T lymphocyte response in milk was of similar or greater magnitude than that in blood and the vaginal tract but higher than that in the colon. Furthermore, the vaccine-elicited SIV Gag-specific CD4(+) and CD8(+) T lymphocyte polyfunctional cytokine responses were more robust in milk than in blood after each virus vector boost. Finally, SIV envelope-specific IgG responses were detected in milk of all monkeys after vaccination, whereas an SIV envelope-specific IgA response was only detected in one vaccinated monkey. Importantly, only limited and transient increases in the proportion of activated or CCR5-expressing CD4(+) T lymphocytes in milk occurred after vaccination. Therefore, systemic DNA prime and virus vector boost of lactating rhesus monkeys elicits potent virus-specific cellular and humoral immune responses in milk and may warrant further investigation as a strategy to impede breast milk transmission of HIV.
Collapse
Affiliation(s)
- Andrew B Wilks
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Edmonds TG, Ding H, Yuan X, Wei Q, Smith KS, Conway JA, Wieczorek L, Brown B, Polonis V, West JT, Montefiori DC, Kappes JC, Ochsenbauer C. Replication competent molecular clones of HIV-1 expressing Renilla luciferase facilitate the analysis of antibody inhibition in PBMC. Virology 2010; 408:1-13. [PMID: 20863545 DOI: 10.1016/j.virol.2010.08.028] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 07/28/2010] [Accepted: 08/22/2010] [Indexed: 01/28/2023]
Abstract
Effective vaccine development for human immunodeficiency virus type 1 (HIV-1) will require assays that ascertain the capacity of vaccine immunogens to elicit neutralizing antibodies (NAb) to diverse HIV-1 strains. To facilitate NAb assessment in peripheral blood mononuclear cell (PBMC)-based assays, we developed an assay-adaptable platform based on a Renilla luciferase (LucR) expressing HIV-1 proviral backbone. LucR was inserted into pNL4-3 DNA, preserving all viral open reading frames. The proviral genome was engineered to facilitate expression of diverse HIV-1 env sequences, allowing analysis in an isogenic background. The resulting Env-IMC-LucR viruses are infectious, and LucR is stably expressed over multiple replications in PBMC. HIV-1 neutralization, targeting TZM-bl cells, was highly correlative comparing virus (LucR) and cell (firefly luciferase) readouts. In PBMC, NAb activity can be analyzed either within a single or multiple cycles of replication. These results represent advancement toward a standardizable PBMC-based neutralization assay for assessing HIV-1 vaccine immunogen efficacy.
Collapse
Affiliation(s)
- Tara G Edmonds
- Department of Molecular and Cellular Pathology, University of Alabama at Birmingham, 701 19th Street South, Birmingham, AL 35294, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Impact of ETIF deletion on safety and immunogenicity of equine herpesvirus type 1-vectored vaccines. J Virol 2010; 84:11602-13. [PMID: 20826695 DOI: 10.1128/jvi.00677-10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Heterologous gene transfer by viral vector systems is often limited by factors such as preexisting immunity, toxicity, low packaging capacity, or weak immunogenic potential. A novel viral vector system derived from equine herpesvirus type 1 (EHV-1) not only overcomes some of these obstacles but also promotes the robust expression of a delivered transgene and the induction of antigen-specific immune responses. Regarding an enhanced safety profile, we assessed the impact of the gene encoding the sole essential tegument protein, ETIF, on the replication and immunogenicity of recombinant EHVs. The deletion of ETIF severely attenuates replication in permissive RK13 cells and a human lung epithelial cell line but without influencing transgene expression. Whereas the intranasal administration of a recombinant luciferase EHV in BALB/c mice resulted in transgene expression in nasal cavities and lungs for 5 to 6 days, the ETIF deletion limited expression to 2 days and resulted in 30-fold-less luminescence. Attenuated replication was accompanied by a decreased capacity to induce CD8(+) T cells against a delivered HIV Gag transgene in BALB/c mice following repeated intranasal application. However, a single subcutaneous immunization with a gag DNA vaccine primed specific T cells for substantial expansion by two subsequent intranasal booster immunizations with either the gag recombinant ETIF mutant or the parental virus. In addition to inducing Gag-specific serum antibodies, this prime-boost strategy clearly outperformed three sequential immunizations with the parental or EHV-ΔETIF virus or repeated DNA vaccination by inducing substantial specific secretory IgA (sIgA) titers.
Collapse
|
21
|
Wei CJ, Boyington JC, McTamney PM, Kong WP, Pearce MB, Xu L, Andersen H, Rao S, Tumpey TM, Yang ZY, Nabel GJ. Induction of broadly neutralizing H1N1 influenza antibodies by vaccination. Science 2010; 329:1060-4. [PMID: 20647428 DOI: 10.1126/science.1192517] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The rapid dissemination of the 2009 pandemic influenza virus underscores the need for universal influenza vaccines that elicit protective immunity to diverse viral strains. Here, we show that vaccination with plasmid DNA encoding H1N1 influenza hemagglutinin (HA) and boosting with seasonal vaccine or replication-defective adenovirus 5 vector encoding HA stimulated the production of broadly neutralizing influenza antibodies. This prime/boost combination increased the neutralization of diverse H1N1 strains dating from 1934 to 2007 as compared to either component alone and conferred protection against divergent H1N1 viruses in mice and ferrets. These antibodies were directed to the conserved stem region of HA and were also elicited in nonhuman primates. Cross-neutralization of H1N1 subtypes elicited by this approach provides a basis for the development of a universal influenza vaccine for humans.
Collapse
Affiliation(s)
- Chih-Jen Wei
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892-3005, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Licensed vaccines against viral diseases generate antibodies that neutralize the infecting virus and protect against infection or disease. Similarly, an effective vaccine against HIV-1 will likely need to induce antibodies that prevent initial infection of host cells or that limit early events of viral dissemination. Such antibodies must target the surface envelope glycoproteins of HIV-1, which are highly variable in sequence and structure. The first subunit vaccines to enter clinical trails were safe and immunogenic but unable to elicit antibodies that neutralized most circulating strains of HIV-1. However, potent virus neutralizing antibodies (NAbs) can develop during the course of HIV-1 infection, and this is the type of antibody response that researchers seek to generate with a vaccine. Thus, current vaccine design efforts have focused on a more detailed understanding of these broadly neutralizing antibodies and their epitopes to inform the design of improved vaccines.
Collapse
Affiliation(s)
- John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
23
|
Rational antibody-based HIV-1 vaccine design: current approaches and future directions. Curr Opin Immunol 2010; 22:358-66. [PMID: 20299194 DOI: 10.1016/j.coi.2010.02.012] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 02/17/2010] [Indexed: 10/19/2022]
Abstract
Many antiviral vaccines elicit neutralizing antibodies as a correlate of protection. For HIV, given the huge variability of the virus, it is widely believed that the induction of a broadly neutralizing antibody (bNAb) response will be crucial in a successful vaccine against the virus. Unfortunately, despite many efforts, the development of an immunogen that elicits bNAbs remains elusive. However, recent structural studies of HIV-1 Env proteins, generation of novel bNAbs, maturation of technologies for the isolation of further antibodies, insights into the requirements for antibody-mediated protection, and novel vaccination approaches are providing grounds for renewed optimism.
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW This review summarizes current and novel virologic reagents employed for the development and application of in-vitro assays that assess neutralizing activity of antibodies against HIV-1. Characteristics of several virologic approaches are placed in context with various cellular targets and assay read-outs intended to determine potency and breadth of neutralization in patient cohorts and clinical vaccine trials. RECENT FINDINGS New molecular virologic reagents developed for in-vitro primary cell-based assays promise to facilitate rigorous and standardized assessment of anti-HIV-1-neutralizing antibody responses elicited by vaccine immunogens. SUMMARY Comprehensive assessment of anti-HIV-1 antibody potency and breadth is essential for evaluating vaccine immunogens, the advancement of vaccine candidates into clinical trials, and ultimately the development of effective vaccine strategies. Env-pseudovirion and recombinant reporter cell line neutralization assays are important tools for rapid and standardized measurement of neutralizing antibody activity. However, recent studies indicate that reporter cell lines fail to detect neutralization activity of certain antibodies observed when analyzed in peripheral blood mononuclear cells and may yield results on neutralizing antibody breadth that are discordant with peripheral blood mononuclear cell assays. Importantly, it remains unknown whether current in-vitro assays may be predictive of a protective neutralizing antibody response elicited by vaccine immunogens. This situation underscores the significance of standardizing existing, complementary methods as well as developing new assay concepts that assess neutralization in primary cells. Thus, this chapter focuses on new virologic reagents that promise to facilitate reaching this goal.
Collapse
|
25
|
Yashima S, Yoshizaki S, Shinoda K, Yoshida A, Kondo A, Mizuguchi H, Ryo A, Okuda K, Shimada M. Co-administration of viral vector-based vaccines suppresses antigen-specific effector CD8 T cells. Vaccine 2010; 28:3257-64. [PMID: 20188679 DOI: 10.1016/j.vaccine.2010.01.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Revised: 01/25/2010] [Accepted: 01/28/2010] [Indexed: 11/18/2022]
Abstract
In this study, we explored immune responses after intramuscular co-administration of the HIV-1 gp160 Env gene-expressing adenovirus (Ad) vector and modified vaccinia virus Ankara (MVA) vector in a mouse model. Surprisingly, the simultaneous vaccination of the two vaccines, either as a mixture or separately, suppressed responses, when compared with the administration of each vaccine separately. Ad vaccine or MVA vaccine, co-administered with a mock MVA or mock Ad vector, also resulted in suppressing HIV-specific effector T-cell responses, and a part of antigen-specific memory T-cell responses. In an in vitro experiment, the two vectors infected individual cells and MVA suppressed the transgene expression produced by the adenovirus vector. This viral interference may involve soluble factor(s), secreted by virus-infected cells. Our study may help in designing a vaccination regimen and in investigating viral interference.
Collapse
Affiliation(s)
- Sayaka Yashima
- Department of Molecular Biodefence Research, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Vaine M, Lu S, Wang S. Progress on the induction of neutralizing antibodies against HIV type 1 (HIV-1). BioDrugs 2009; 23:137-53. [PMID: 19627166 DOI: 10.2165/00063030-200923030-00001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Infection with HIV type 1 (HIV-1), the causative agent of AIDS, is one of the most catastrophic pandemics to affect human healthcare in the latter 20th century. The best hope of controlling this pandemic is the development of a successful prophylactic vaccine. However, to date, this goal has proven to be exceptionally elusive. The recent failure of an experimental vaccine in a phase IIb study, named the STEP trial, intended solely to elicit cell-mediated immune responses against HIV-1, has highlighted the need for a balanced immune response consisting of not only cellular immunity but also a broad and potent humoral antibody response that can prevent infection with HIV-1. This article reviews the efforts made up to this point to elicit such antibody responses, especially with regard to the use of a DNA prime-protein boost regimen, which has been proven to be a highly effective platform for the induction of neutralizing antibodies in both animal and early-phase human studies.
Collapse
Affiliation(s)
- Michael Vaine
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | |
Collapse
|
27
|
Santra S, Sun Y, Korioth-Schmitz B, Fitzgerald J, Charbonneau C, Santos G, Seaman MS, Ratcliffe SJ, Montefiori DC, Nabel GJ, Ertl HCJ, Letvin NL. Heterologous prime/boost immunizations of rhesus monkeys using chimpanzee adenovirus vectors. Vaccine 2009; 27:5837-45. [PMID: 19660588 PMCID: PMC2955883 DOI: 10.1016/j.vaccine.2009.07.050] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 07/16/2009] [Accepted: 07/18/2009] [Indexed: 11/28/2022]
Abstract
Pre-existing immunity to human adenovirus serotype 5 (AdHu5) has been shown to suppress the immunogenicity of recombinant Ad5 (rAdHu5) vector-based vaccines for human immunodeficiency virus type 1 (HIV-1) in both preclinical studies and clinical trials. As a potential solution to this problem we developed adenovirus vaccine vectors of chimpanzee origin. In the present study we assessed the immunogenicity of various chimpanzee adenovirus vectors in a prime/boost regimen to HIV-1 envelope and SIV Gag-Pol in rhesus monkeys and their ability to protect against pathogenic viral challenge. Although rAdHu5-primed monkeys had higher magnitude T cell responses than rAdC7 or rAdC68 prior to challenge, the rAdC7-rAdC1/C5 and rAdHu5-rAdC1/C5 immunizations resulted in comparable magnitude recall cellular immune responses and comparable level of control of viremia post-challenge.
Collapse
Affiliation(s)
- Sampa Santra
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Yue Sun
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Birgit Korioth-Schmitz
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Julie Fitzgerald
- Wistar Institute, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Cherie Charbonneau
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Giannina Santos
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Michael S. Seaman
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Sarah J. Ratcliffe
- Wistar Institute, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - David C. Montefiori
- Department of Surgery, Laboratory for AIDS Vaccine Research and Development, Duke University Medical Center, Durham, NC
| | - Gary J. Nabel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Hildegund C. J. Ertl
- Wistar Institute, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Norman L. Letvin
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
28
|
A comparative study of HIV-1 clade C env evolution in a Zambian infant with an infected rhesus macaque during disease progression. AIDS 2009; 23:1817-28. [PMID: 19609201 DOI: 10.1097/qad.0b013e32832f3da6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate whether HIV-1 clade C (HIV-C) envelope variations that arise during disease progression in rhesus macaque model reflect changes that occur naturally in human infection. DESIGN An infant macaque was infected with SHIV-1157i, an R5 tropic clade C SHIV, that expresses a primary HIV-C envelope derived from an infected human infant and monitored over a 5-year period. Genetic variation of the V1-V5 envelope region, which is the main target for humoral immune responses, derived from the infected macaque and infant was examined. METHODS The V1-V5 envelope region was cloned and sequenced from longitudinal peripheral blood mononuclear cell samples collected from the infected macaque and infant. Phylogenetic analysis [phylogenetic tree, diversity, divergence, ratio of nonsynonymous (dN) and synonymous substitution (dS) and dN distribution] was performed. Plasma RNA viral load, CD4(+) T-cell count, changes in the length of V1-V5 region, putative N-linked glycosylation site number and distribution were also measured. RESULTS Phylogenetic analysis revealed that changes in the macaque closely reflected those of the infant during disease progression. Similar distribution patterns of dN and hot spots were observed between the macaque and infant. Analysis of putative N-linked glycosylation sites revealed several common variations between the virus populations in the two host species. These variations correlate with decline of CD4 T-cell count in the macaque and might be linked with disease progression. CONCLUSION SHIV-C infection of macaque is a relevant animal model for studying variation of primary HIV-C envelope during disease progression and could be used to analyze the selection pressures that are associated with those changes.
Collapse
|
29
|
A recombinant adenovirus prime-virus-like particle boost regimen elicits effective and specific immunities against norovirus in mice. Vaccine 2009; 27:5233-8. [DOI: 10.1016/j.vaccine.2009.06.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 06/11/2009] [Accepted: 06/18/2009] [Indexed: 11/19/2022]
|
30
|
Zolla-Pazner S, Cohen S, Pinter A, Krachmarov C, Wrin T, Wang S, Lu S. Cross-clade neutralizing antibodies against HIV-1 induced in rabbits by focusing the immune response on a neutralizing epitope. Virology 2009; 392:82-93. [PMID: 19632700 DOI: 10.1016/j.virol.2009.05.039] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 05/18/2009] [Accepted: 05/28/2009] [Indexed: 11/17/2022]
Abstract
Studies were performed to induce cross-clade neutralizing antibodies (Abs) by testing various combinations of prime and boost constructs that focus the immune response on structurally-conserved epitopes in the V3 loop of HIV-1 gp120. Rabbits were immunized with gp120 DNA containing a V3 loop characterized by the GPGR motif at its tip, and/or with gp120 DNA with a V3 loop carrying the GPGQ motif. Priming was followed by boosts with V3-fusion proteins (V3-FPs) carrying the V3 sequence from a subtype B virus (GPGR motif), and/or with V3 sequences from subtypes A and C (GPGQ motif). The broadest and most consistent neutralizing responses were generated when using a clade C gp120 DNA prime and with the V3(B)-FP boost. Immune sera displayed neutralizing activity in three assays against pseudoviruses and primary isolates from subtypes A, AG, B, C, and D. Polyclonal Abs in the immune rabbit sera neutralized viruses that were not neutralized by pools of human anti-V3 monoclonal Abs. Greater than 80% of the neutralizing Abs were specific for V3, showing that the immune response could be focused on a neutralizing epitope and that vaccine-induced anti-V3 Abs have cross-clade neutralizing activity.
Collapse
Affiliation(s)
- Susan Zolla-Pazner
- New York University School of Medicine, 550 First Avenue, New York, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Haut LH, Ertl HCJ. Obstacles to the successful development of an efficacious T cell-inducing HIV-1 vaccine. J Leukoc Biol 2009; 86:779-93. [PMID: 19597003 DOI: 10.1189/jlb.0209094] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
An efficacious vaccine to HIV-1 is direly needed to stem the global pandemic. Immunogens that elicit broadly cross-neutralizing antibodies to HIV-1 remain elusive, and thus, most HIV-1 vaccine efforts are focusing on induction of T cells. The notion that T cells can mediate protection against HIV-1 has been called into question by the failure of the STEP trial, which was designed to test this concept by the use of an E1-deleted Ad vaccine carrier. Lack of efficacy of the STEP trial vaccine underscores our limited knowledge about correlates of immune protection against HIV-1 and stresses the need for an enhanced commitment to basic research, including preclinical and clinical vaccine studies. In this review, we discuss known correlates of protection against HIV-1 and different vaccine strategies that have been or are being explored to induce such correlates, focusing on T cell-inducing vaccines and particularly on Ad vectors.
Collapse
Affiliation(s)
- Larissa Herkenhoff Haut
- Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | | |
Collapse
|
32
|
Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. J Virol 2009; 83:7337-48. [PMID: 19439467 DOI: 10.1128/jvi.00110-09] [Citation(s) in RCA: 466] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of a rapid and efficient system to identify human immunodeficiency virus type 1 (HIV-1)-infected individuals with broad and potent HIV-1-specific neutralizing antibody responses is an important step toward the discovery of critical neutralization targets for rational AIDS vaccine design. In this study, samples from HIV-1-infected volunteers from diverse epidemiological regions were screened for neutralization responses using pseudovirus panels composed of clades A, B, C, and D and circulating recombinant forms (CRFs). Initially, 463 serum and plasma samples from Australia, Rwanda, Uganda, the United Kingdom, and Zambia were screened to explore neutralization patterns and selection ranking algorithms. Samples were identified that neutralized representative isolates from at least four clade/CRF groups with titers above prespecified thresholds and ranked based on a weighted average of their log-transformed neutralization titers. Linear regression methods selected a five-pseudovirus subset, representing clades A, B, and C and one CRF01_AE, that could identify top-ranking samples with 50% inhibitory concentration (IC(50)) neutralization titers of >or=100 to multiple isolates within at least four clade groups. This reduced panel was then used to screen 1,234 new samples from the Ivory Coast, Kenya, South Africa, Thailand, and the United States, and 1% were identified as elite neutralizers. Elite activity is defined as the ability to neutralize, on average, more than one pseudovirus at an IC(50) titer of 300 within a clade group and across at least four clade groups. These elite neutralizers provide promising starting material for the isolation of broadly neutralizing monoclonal antibodies to assist in HIV-1 vaccine design.
Collapse
|
33
|
Pahar B, Lackner AA, Piatak M, Lifson JD, Wang X, Das A, Ling B, Montefiori DC, Veazey RS. Control of viremia and maintenance of intestinal CD4(+) memory T cells in SHIV(162P3) infected macaques after pathogenic SIV(MAC251) challenge. Virology 2009; 387:273-84. [PMID: 19298994 PMCID: PMC2674129 DOI: 10.1016/j.virol.2009.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 12/17/2008] [Accepted: 02/06/2009] [Indexed: 11/16/2022]
Abstract
Recent HIV vaccine failures have prompted calls for more preclinical vaccine testing in non-human primates. However, similar to HIV infection of humans, developing a vaccine that protects macaques from infection following pathogenic SIV(MAC251) challenge has proven difficult, and current vaccine candidates at best, only reduce viral loads after infection. Here we demonstrate that prior infection with a chimeric simian-human immunodeficiency virus (SHIV) containing an HIV envelope gene confers protection against intravenous infection with the heterologous, highly pathogenic SIV(MAC251) in rhesus macaques. Although definitive immune correlates of protection were not identified, preservation and/or restoration of intestinal CD4(+) memory T cells were associated with protection from challenge and control of viremia. These results suggest that protection against pathogenic lentiviral infection or disease progression is indeed possible, and may correlate with preservation of mucosal CD4(+) T cells.
Collapse
Affiliation(s)
- Bapi Pahar
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Recombinant Mycobacterium bovis BCG prime-recombinant adenovirus boost vaccination in rhesus monkeys elicits robust polyfunctional simian immunodeficiency virus-specific T-cell responses. J Virol 2009; 83:5505-13. [PMID: 19297477 DOI: 10.1128/jvi.02544-08] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
While mycobacteria have been proposed as vaccine vectors because of their persistence and safety, little has been done systematically to optimize their immunogenicity in nonhuman primates. We successfully generated recombinant Mycobacterium bovis BCG (rBCG) expressing simian immunodeficiency virus (SIV) Gag and Pol as multigenic, nonintegrating vectors, but rBCG-expressing SIV Env was unstable. A dose and route determination study in rhesus monkeys revealed that intramuscular administration of rBCG was associated with local reactogenicity, whereas intravenous and intradermal administration of 10(6) to 10(8) CFU of rBCG was well tolerated. After single or repeat rBCG inoculations, monkeys developed high-frequency gamma interferon enzyme-linked immunospot responses against BCG purified protein derivative. However, the same animals developed only modest SIV-specific CD8(+) T-cell responses. Nevertheless, high-frequency SIV-specific cellular responses were observed in the rBCG-primed monkeys after boosting with recombinant adenovirus 5 (rAd5) expressing the SIV antigens. These cellular responses were of greater magnitude and more persistent than those generated after vaccination with rAd5 alone. The vaccine-elicited cellular responses were predominantly polyfunctional CD8(+) T cells. These findings support the further exploration of mycobacteria as priming vaccine vectors.
Collapse
|
35
|
Abstract
We have focused our research on understanding the basic biology of and developing novel therapeutic and prophylactic DNA vaccines. We have among others three distinct primary areas of interest which include: 1. Enhancing in vivo delivery and transfection of DNA vaccine vectors 2. Improving DNA vaccine construct immunogenicity 3. Using molecular adjuvants to modulate and skew immune responses. Key to the immunogenicity of DNA vaccines is the presentation of expressed antigen to antigen-presenting cells. To improve expression and presentation of antigen, we have investigated various immunization methods with current focus on a combination of intramuscular injection and electroporation. To improve our vaccine constructs, we also employed methods such as RNA/codon optimization and antigen consensus to enhance expression and cellular/humoral cross-reactivity, respectively. Our lab also researches the potential of various molecular adjuvants to skew Th1/Th2 responses, enhance cellular/humoral responses, and improve protection in various animal models. Through improving our understanding of basic immunology as it is related to DNA vaccine technology, our goal is to develop the technology to the point of utility for human and animal health.
Collapse
Affiliation(s)
- Shaheed A Abdulhaqq
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, 505 Stellar-Chance Laboratories, Philadelphia, PA, 19104, USA
| | | |
Collapse
|
36
|
Pozzi E, Zanotto C, Pacchioni S, De Giuli Morghen C, Radaelli A. MHC-restricted cytotoxic T-lymphocyte assay: An improved method based on normal and SV40-immortalized rabbit epidermal target cells. J Virol Methods 2009; 155:77-81. [DOI: 10.1016/j.jviromet.2008.09.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 09/19/2008] [Accepted: 09/25/2008] [Indexed: 11/26/2022]
|
37
|
Bogers WMJM, Davis D, Baak I, Kan E, Hofman S, Sun Y, Mortier D, Lian Y, Oostermeijer H, Fagrouch Z, Dubbes R, van der Maas M, Mooij P, Koopman G, Verschoor E, Langedijk JPM, Zhao J, Brocca-Cofano E, Robert-Guroff M, Srivastava I, Barnett S, Heeney JL. Systemic neutralizing antibodies induced by long interval mucosally primed systemically boosted immunization correlate with protection from mucosal SHIV challenge. Virology 2008; 382:217-25. [PMID: 18947849 PMCID: PMC2723753 DOI: 10.1016/j.virol.2008.09.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 08/12/2008] [Accepted: 09/17/2008] [Indexed: 01/11/2023]
Abstract
Immune correlates of vaccine protection from HIV-1 infection would provide important milestones to guide HIV-1 vaccine development. In a proof of concept study using mucosal priming and systemic boosting, the titer of neutralizing antibodies in sera was found to correlate with protection of mucosally exposed rhesus macaques from SHIV infection. Mucosal priming consisted of two sequential immunizations at 12-week intervals with replicating host range mutants of adenovirus type 5 (Ad5hr) expressing the HIV-1(89.6p) env gene. Following boosting with either heterologous recombinant protein or alphavirus replicons at 12-week intervals animals were intrarectally exposed to infectious doses of the CCR5 tropic SHIV(SF162p4). Heterologous mucosal prime systemic boost immunization elicited neutralizing antibodies (Nabs), antibody-dependent cytotoxicity (ADCC), and specific patterns of antibody binding to envelope peptides. Vaccine induced protection did not correlate with the type of boost nor T-cell responses, but rather with the Nab titer prior to exposure.
Collapse
Affiliation(s)
- Willy M J M Bogers
- Department of Virology, Biomedical Primate Research Centre (BPRC), P.O. Box 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The best hope of controlling the HIV pandemic is the development of an effective vaccine. In addition to the stimulation of virus neutralising antibodies, a vaccine will need an effective T-cell response against the virus. Vaccines based on recombinant adenoviruses (rAd) are promising candidates to stimulate anti-HIV T-cell responses. This review discusses the different rAd vector types, problems raised by host immune responses against them and strategies that are being adopted to overcome this problem. Vaccines need to target and stimulate dendritic cells and thus the tropism and interaction of rAd-based vaccines with these cells is covered. Different rAd vaccination regimes and the need to stimulate mucosal responses are discussed together with data from animal studies on immunogenicity and virus challenge experiments. The review ends with a discussion of the recent disappointing Merck HIV vaccine trial.
Collapse
Affiliation(s)
- Steven Patterson
- Department of Immunology, Imperial College, Chelsea and Westminster Hospital, London, UK.
| | | | | |
Collapse
|
39
|
Du Y, Dai J, Li Y, Li C, Qi J, Duan S, Jiang P. Immune responses of recombinant adenovirus co-expressing VP1 of foot-and-mouth disease virus and porcine interferon α in mice and guinea pigs. Vet Immunol Immunopathol 2008; 124:274-83. [DOI: 10.1016/j.vetimm.2008.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2007] [Revised: 01/28/2008] [Accepted: 04/09/2008] [Indexed: 10/22/2022]
|
40
|
Du Y, Li Y, He H, Qi J, Jiang W, Wang X, Tang B, Cao J, Wang X, Jiang P. Enhanced immunogenicity of multiple-epitopes of foot-and-mouth disease virus fused with porcine interferon α in mice and protective efficacy in guinea pigs and swine. J Virol Methods 2008; 149:144-52. [DOI: 10.1016/j.jviromet.2007.12.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 12/09/2007] [Accepted: 12/20/2007] [Indexed: 11/26/2022]
|
41
|
Pan Q, He K, Huang K. Development of recombinant porcine parvovirus-like particles as an antigen carrier formed by the hybrid VP2 protein carrying immunoreactive epitope of porcine circovirus type 2. Vaccine 2008; 26:2119-26. [PMID: 18378364 DOI: 10.1016/j.vaccine.2008.02.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2007] [Revised: 01/01/2008] [Accepted: 02/10/2008] [Indexed: 11/16/2022]
Abstract
Virus-like particles (VLPs) are non-replicative vectors for delivery of heterologous epitopes and induction of immune responses. In this study, a self-assembled porcine parvovirus (PPV) VP2 capsid protein [PPV:VLP-(PCV2)] carrying immunoreactive epitopes, residues 165-200 from the Porcine circovirus 2 (PCV2) virus nucleoprotein was constructed. Immunogenicity study was carried out with hybrid VLPs derived from HEK-293 cells infected with recombinant adenovirus vectors. To our knowledge, this study presents the first demonstration that hybrid non-replicative PPV VLPs carrying PCV2 immunoreactive epitopes can induce stronger antibody responses against PCV2 than recombinant adenovirus of PCV2 ORF2, in the absence of any adjuvant. The hybrid VLPs [PPV:VLP-(PCV2)] might be a promising candidate vaccine for better prevention of the diseases associated with PCV2 as well as with co-infection by PCV2 and PPV.
Collapse
Affiliation(s)
- Qunxing Pan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, Jiangsu Province, China
| | | | | |
Collapse
|
42
|
Patterson LJ, Beal J, Demberg T, Florese RH, Malkevich N, Venzon D, Aldrich K, Richardson E, Kalyanaraman VS, Kalisz I, Lee EM, Montefiori DC, Robey FA, Robert-Guroff M. Replicating adenovirus HIV/SIV recombinant priming alone or in combination with a gp140 protein boost results in significant control of viremia following a SHIV89.6P challenge in Mamu-A*01 negative rhesus macaques. Virology 2008; 374:322-37. [PMID: 18252262 DOI: 10.1016/j.virol.2007.12.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 11/13/2007] [Accepted: 12/24/2007] [Indexed: 11/27/2022]
Abstract
Previously, replicating adenovirus type 5 host range (Ad5hr)-HIV/SIV recombinant priming in combination with SIV envelope boosting, resulted in significant, durable protection in 39% of rhesus macaques after SIVmac251 challenge. Both Env-specific antibody mediating ADCC, and cellular immunity correlated with protection. Here we evaluate the relative immunogenicities of novel HIV proteins and their contribution to protection in a SHIV89.6P model. All groups were primed with Ad-HIVenv89.6P, SIVgag239, and SIVnef239 recombinants. One group was not boosted, one received HIV89.6Pgp140DeltaCFI protein, and one a novel HIV-1 poly-peptide "peptomer". The HIV89.6Pgp140DeltaCFI protein in adjuvant strongly boosted Env-specific antibody and memory T cell responses in blood and tissue, resulting in significant reductions in acute and set point viremia. Macaques not boosted, showed a significant reduction in set point viremia, a full 32 weeks after the last Ad priming immunization. The HIV peptomer-boosted group showed a trend toward chronic viremia reduction, but was not protected.
Collapse
Affiliation(s)
- L Jean Patterson
- Vaccine Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bielinska AU, Janczak KW, Landers JJ, Markovitz DM, Montefiori DC, Baker JR. Nasal immunization with a recombinant HIV gp120 and nanoemulsion adjuvant produces Th1 polarized responses and neutralizing antibodies to primary HIV type 1 isolates. AIDS Res Hum Retroviruses 2008; 24:271-81. [PMID: 18260780 DOI: 10.1089/aid.2007.0148] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epidemiological and experimental data suggest that both robust neutralizing antibodies and potent cellular responses play important roles in controlling primary HIV-1 infection. In this study we have investigated the induction of systemic and mucosal immune responses to HIV gp120 monomer immunogen administered intranasally in a novel, oil-in-water nanoemulsion (NE) adjuvant. Mice and guinea pigs intranasally immunized by the application of recombinant HIV gp120 antigen mixed in NE demonstrated robust serum anti-gp120 IgG, as well as bronchial, vaginal, and serum anti-gp120 IgA in mice. The serum of these animals demonstrated antibodies that cross-reacted with heterologous serotypes of gp120 and had significant neutralizing activity against two clade-B laboratory strains of HIV (HIVBaL and HIVSF162) and five primary HIV-1 isolates. The analysis of gp120-specific CTL proliferation, INF-gamma induction, and prevalence of anti-gp120 IgG2 subclass antibodies indicated that nasal vaccination in NE also induced systemic, Th1-polarized cellular immune responses. This study suggests that NE should be evaluated as a mucosal adjuvant for multivalent HIV vaccines.
Collapse
Affiliation(s)
- Anna U. Bielinska
- Michigan Nanotechnology Institute for Medicine and Biological Sciences (MNIMBS), University of Michigan, Ann Arbor, Michigan 48109
| | - Katarzyna W. Janczak
- Michigan Nanotechnology Institute for Medicine and Biological Sciences (MNIMBS), University of Michigan, Ann Arbor, Michigan 48109
| | - Jeffrey J. Landers
- Michigan Nanotechnology Institute for Medicine and Biological Sciences (MNIMBS), University of Michigan, Ann Arbor, Michigan 48109
| | - David M. Markovitz
- Internal Medicine, Infectious Diseases, University of Michigan, Ann Arbor, Michigan 48109
| | - David C. Montefiori
- Department of Surgery, Laboratory for AIDS Vaccine Research and Development, Duke University Medical Center, Durham, North Carolina 27706
| | - James R. Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences (MNIMBS), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
44
|
Koopman G, Mortier D, Hofman S, Mathy N, Koutsoukos M, Ertl P, Overend P, van Wely C, Thomsen LL, Wahren B, Voss G, Heeney JL. Immune-response profiles induced by human immunodeficiency virus type 1 vaccine DNA, protein or mixed-modality immunization: increased protection from pathogenic simian–human immunodeficiency virus viraemia with protein/DNA combination. J Gen Virol 2008; 89:540-5533. [PMID: 18198386 DOI: 10.1099/vir.0.83384-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Current data suggest that prophylactic human immunodeficiency virus type 1 (HIV) vaccines will be most efficacious if they elicit a combination of adaptive humoral and T-cell responses. Here, we explored the use of different vaccine strategies in heterologous prime–boost regimes and evaluated the breadth and nature of immune responses in rhesus monkeys induced by epidermally delivered plasmid DNA or recombinant HIV proteins formulated in the AS02A adjuvant system. These immunogens were administered alone or as either prime or boost in mixed-modality regimes. DNA immunization alone induced cell-mediated immune (CMI) responses, with a strong bias towards Th1-type cytokines, and no detectable antibodies to the vaccine antigens. Whenever adjuvanted protein was used as a vaccine, either alone or in a regime combined with DNA, high-titre antibody responses to all vaccine antigens were detected in addition to strong Th1- and Th2-type CMI responses. As the vaccine antigens included HIV-1 Env, Nef and Tat, as well as simian immunodeficiency virus (SIV)mac239 Nef, the animals were subsequently exposed to a heterologous, pathogenic simian–human immunodeficiency virus (SHIV)89.6p challenge. Protection against sustained high virus load was observed to some degree in all vaccinated groups. Suppression of virus replication to levels below detection was observed most frequently in the group immunized with protein followed by DNA immunization, and similarly in the group immunized with DNA alone. Interestingly, control of virus replication was associated with increased SIV Nef- and Gag-specific gamma interferon responses observed immediately following challenge.
Collapse
MESH Headings
- AIDS Vaccines/immunology
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/pharmacology
- Gene Products, env/genetics
- Gene Products, env/immunology
- Gene Products, env/metabolism
- Gene Products, tat/genetics
- Gene Products, tat/immunology
- Gene Products, tat/metabolism
- HIV/genetics
- HIV/immunology
- HIV/metabolism
- Human Immunodeficiency Virus Proteins/administration & dosage
- Human Immunodeficiency Virus Proteins/genetics
- Humans
- Immunization
- Macaca mulatta
- Simian Immunodeficiency Virus/physiology
- Vaccines, DNA/immunology
- Vaccines, Subunit/immunology
- Viremia
- Virus Replication
Collapse
Affiliation(s)
- Gerrit Koopman
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| | - Sam Hofman
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| | | | | | - Peter Ertl
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Phil Overend
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Cathy van Wely
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Lindy L Thomsen
- GlaxoSmithKline Biopharmaceuticals CEDD Biology, Stevenage, UK
| | - Britta Wahren
- Swedish Institute for Infectious Disease Control, Karolinska Institutet, Stockholm, Sweden
| | - Gerald Voss
- GlaxoSmithKline Biologicals, Rixensart, Belgium
| | - Jonathan L Heeney
- Department of Veterinary Medicine, University of Cambridge, UK
- Department of Virology, Biomedical Primate Research Center (BPRC), 2288 GH Rijswijk, The Netherlands
| |
Collapse
|
45
|
Li Y, Cleveland B, Klots I, Travis B, Richardson BA, Anderson D, Montefiori D, Polacino P, Hu SL. Removal of a single N-linked glycan in human immunodeficiency virus type 1 gp120 results in an enhanced ability to induce neutralizing antibody responses. J Virol 2008; 82:638-51. [PMID: 17959660 PMCID: PMC2224603 DOI: 10.1128/jvi.01691-07] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Accepted: 10/18/2007] [Indexed: 11/20/2022] Open
Abstract
Glycans on human immunodeficiency virus (HIV) envelope protein play an important role in infection and evasion from host immune responses. To examine the role of specific glycans, we introduced single or multiple mutations into potential N-linked glycosylation sites in hypervariable regions (V1 to V3) of the env gene of HIV type 1 (HIV-1) 89.6. Three mutants tested showed enhanced sensitivity to soluble CD4. Mutant N7 (N197Q) in the carboxy-terminal stem of the V2 loop showed the most pronounced increase in sensitivity to broadly neutralizing antibodies (NtAbs), including those targeting the CD4-binding site (IgG1b12) and the V3 loop (447-52D). This mutant is also sensitive to CD4-induced NtAb 17b in the absence of CD4. Unlike the wild-type (WT) Env, mutant N7 mediates CD4-independent infection in U87-CXCR4 cells. To study the immunogenicity of mutant Env, we immunized pig-tailed macaques with recombinant vaccinia viruses, one expressing SIVmac239 Gag-Pol and the other expressing HIV-1 89.6 Env gp160 in WT or mutant forms. Animals were boosted 14 to 16 months later with simian immunodeficiency virus gag DNA and the cognate gp140 protein before intrarectal challenge with SHIV89.6P-MN. Day-of-challenge sera from animals immunized with mutant N7 Env had significantly higher and broader neutralizing activities than sera from WT Env-immunized animals. Neutralizing activity was observed against SHIV89.6, SHIV89.6P-MN, HIV-1 SF162, and a panel of subtype B primary isolates. Compared to control animals, immunized animals showed significant reduction of plasma viral load and increased survival after challenge, which correlated with prechallenge NtAb titers. These results indicate the potential advantages for glycan modification in vaccine design, although the role of specific glycans requires further examination.
Collapse
Affiliation(s)
- Yun Li
- Washington National Primate Center, University of Washington, 3000 Western Ave., Seattle, WA 98121, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Xu HM, Jiang H, Wang J, Luo B, Xie JX. Over-expressed human divalent metal transporter 1 is involved in iron accumulation in MES23.5 cells. Neurochem Int 2007; 52:1044-51. [PMID: 18082289 DOI: 10.1016/j.neuint.2007.10.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 10/02/2007] [Accepted: 10/19/2007] [Indexed: 11/29/2022]
Abstract
Elevated iron accumulation has been reported in brain regions in some neurodegenerative disorders. However, the mechanism for this is largely unknown. Divalent metal transporter 1 (DMT1) is an important divalent cation transporter. The aim of the present study is to construct recombinant adenovirus encoding human DMT1 with iron responsive element (DMT1+IRE) and infect MES23.5 dopaminergic cells in order to investigate the relationship between increased DMT1+IRE expression and iron accumulation. The human DMT1 gene was obtained by RT-PCR from tissues of human duodenum. AdDMT1+IRE was successfully constructed and identified by PCR, restriction endonuclease analyses and DNA sequencing, respectively. It was able to efficiently infect MES23.5 cells, which was confirmed by RT-PCR and Western blots. When incubated with 100 microM ferrous iron for 6h, the intracellular iron levels dramatically increased in AdDMT1+IRE infected MES23.5 cells compared to the solely adenovirus infected cells. Meanwhile, the levels of hydroxyl free radicals and malondialdehyde (MDA) in these cells increased. This led to the activation of caspase-3. The apoptosis in AdDMT1+IRE infected cells was shown with hypercondensed nuclei using Hoechst staining. Analysis of DNA extracted from these cells showed the typical "ladder pattern", indicating the formation of mono- and oligonucleosomes. These results suggested that increased DMT1+IRE expression in MES23.5 cells caused the increased intracellular iron accumulation. This resulted in the increased oxidative stress leading to ultimate cell apoptosis.
Collapse
Affiliation(s)
- Hua-Min Xu
- Department of Physiology, Medical College of Qingdao University, No. 308 Ningxia Road, Qingdao, China
| | | | | | | | | |
Collapse
|
47
|
Seaman MS, Leblanc DF, Grandpre LE, Bartman MT, Montefiori DC, Letvin NL, Mascola JR. Standardized assessment of NAb responses elicited in rhesus monkeys immunized with single- or multi-clade HIV-1 envelope immunogens. Virology 2007; 367:175-86. [PMID: 17599382 PMCID: PMC2075526 DOI: 10.1016/j.virol.2007.05.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 04/30/2007] [Accepted: 05/21/2007] [Indexed: 11/26/2022]
Abstract
The genetic diversity of HIV-1 envelope glycoproteins (Env) remains a major obstacle to the development of an antibody-based AIDS vaccine. The present studies examine the breadth and magnitude of neutralizing antibody (NAb) responses in rhesus monkeys after immunization with DNA prime-recombinant adenovirus (rAd) boost vaccines encoding either single or multiple genetically distant Env immunogens, and subsequently challenged with a pathogenic simian-human immunodeficiency virus (SHIV-89.6P). Using a standardized multi-tier panel of reference Env pseudoviruses for NAb assessment, we show that monkeys immunized with a mixture of Env immunogens (clades A, B, and C) exhibited a greater breadth of NAb activity against neutralization-sensitive Tier 1 viruses following both vaccination and challenge compared to monkeys immunized with a single Env immunogen (clade B or C). However, all groups of Env-vaccinated monkeys demonstrated only limited neutralizing activity against Tier 2 pseudoviruses, which are more characteristic of the neutralization sensitivity of circulating HIV-1. Notably, the development of a post-challenge NAb response against SHIV-89.6P was similar in monkeys receiving either clade B, clade C, or clade A+B+C Env immunogens, suggesting cross-clade priming of NAb responses. In addition, vaccines encoding Env immunogens heterologous to SHIV-89.6P primed for a rapid anamnestic NAb response following infection compared to vaccines lacking an Env component. These results show that DNA/rAd immunization with multiple diverse Env immunogens is a viable approach for enhancing the breadth of NAb responses against HIV-1, and suggest that Env immunogens can prime for anamnestic NAb responses against a heterologous challenge virus.
Collapse
Affiliation(s)
- Michael S Seaman
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, 330 Brookline Ave/RE-204, Boston, MA 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Maggiorella MT, Sernicola L, Crostarosa F, Belli R, Pavone-Cossut MR, Macchia I, Farcomeni S, Tenner-Racz K, Racz P, Ensoli B, Titti F. Multiprotein genetic vaccine in the SIV-Macaca animal model: a promising approach to generate sterilizing immunity to HIV infection. J Med Primatol 2007; 36:180-94. [PMID: 17669207 DOI: 10.1111/j.1600-0684.2007.00236.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Vaccine combining structural and regulatory proteins is an emerging approach to develop an HIV/AIDS vaccine and therefore, the immunogenicity and efficacy of two regimens of immunization combining structural (Gag/Pol, Env) and regulatory (Rev, Tat, Nef) Simian immunodeficiency virus (SIV) proteins were compared in cynomolgus monkeys. METHODS Monkeys were immunized with Modified Vaccine Ankara vector (MVA-J5) (protocol 1) or with DNA, Semliki forest virus and MVA vectors (DNA/SFV/MVA) (protocol 2). At week 32, all monkeys were challenge intravenously (protocol 1) or intrarectally (protocol 2) with 50 MID(50) of SIVmac251. Humoral, proliferative responses and in particular in protocol 2, the frequency of IFN-gamma producing cells, were measured in all monkeys before and after the challenge. RESULTS Both vaccine regimens elicited humoral and proliferative responses but failed to induce neutralizing antibodies. Upon intravenous challenge, two out of three MVA-J5 vaccinated monkeys exhibited a long-term control of the viral replication whereas DNA/SFV/MVA vaccine abrogated the virus replication up to undetectable level in three out of four vaccinated monkeys. A major contribution to this vaccine effect appeared to be the IFN-gamma/ELISPOT responses to vaccine antigens (Gag, Rev Tat and Nef). CONCLUSIONS These results, indicate that multiprotein heterologous prime-boost vaccination can induce a robust vaccine-induced immunity able to abrogate virus replication.
Collapse
Affiliation(s)
- Maria Teresa Maggiorella
- Division of Experimental Retrovirology and Non-Human Primate Models, National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Someya K, Xin KQ, Ami Y, Izumi Y, Mizuguchi H, Ohta S, Yamamoto N, Honda M, Okuda K. Chimeric adenovirus type 5/35 vector encoding SIV gag and HIV env genes affords protective immunity against the simian/human immunodeficiency virus in monkeys. Virology 2007; 367:390-7. [PMID: 17628628 DOI: 10.1016/j.virol.2007.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 04/30/2007] [Accepted: 06/14/2007] [Indexed: 10/23/2022]
Abstract
Replication-defective adenovirus type 5 (Ad5) vector-based vaccines are widely known to induce strong immunity against immunodeficiency viruses. To exploit this immunogenicity while overcoming the potential problem of preexisting immunity against human adenoviruses type 5, we developed a recombinant chimeric adenovirus type 5 with type 35 fiber vector (rAd5/35). We initially produced a simian immunodeficiency virus (SIV) gag DNA plasmid (rDNA-Gag), a human immunodeficiency virus type 1 (HIV-1) 89.6 env DNA plasmid (rDNA-Env) and a recombinant Ad5/35 vector encoding the SIV gag and HIV env gene (rAd5/35-Gag and rAd5/35-Env). Prime-boost vaccination with rDNA-Gag and -Env followed by high doses of rAd5/35-Gag and -Env elicited higher levels of cellular immune responses than did rDNAs or rAd5/35s alone. When challenged with a pathogenic simian human immunodeficiency virus (SHIV), animals receiving a prime-boost regimen or rAd5/35s alone maintained a higher number of CD4(+) T cells and remarkably suppressed plasma viral RNA loads. These findings suggest the clinical promise of an rAd5/35 vector-based vaccine.
Collapse
Affiliation(s)
- Kenji Someya
- Department of Virology III, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tavel JA, Martin JE, Kelly GG, Enama ME, Shen JM, Gomez PL, Andrews CA, Koup RA, Bailer RT, Stein JA, Roederer M, Nabel GJ, Graham BS. Safety and immunogenicity of a Gag-Pol candidate HIV-1 DNA vaccine administered by a needle-free device in HIV-1-seronegative subjects. J Acquir Immune Defic Syndr 2007; 44:601-5. [PMID: 17325604 PMCID: PMC2365751 DOI: 10.1097/qai.0b013e3180417cb6] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the safety and immunogenicity of a candidate HIV DNA vaccine administered using a needle-free device. DESIGN In this phase 1, dose escalation, double-blind, placebo-controlled clinical trial, 21 healthy adults were randomized to receive placebo or 0.5, 1.5, or 4 mg of a single plasmid expressing a Gag/Pol fusion protein. Each participant received repeat immunizations at days 28 and 56 after the first inoculation. Safety and immunogenicity data were collected. RESULTS The vaccine was well tolerated, with most adverse events being mild injection site reactions, including pain, tenderness, and erythema. No dose-limiting toxicities occurred. HIV-specific antibody response was not detected in any vaccinee by enzyme-linked immunosorbent assay. HIV-specific T-cell responses to Gag or Pol as measured by enzyme-linked immunospot assay and intracellular cytokine staining were of low frequency and magnitude. CONCLUSIONS This candidate HIV DNA vaccine was safe and well tolerated. No HIV-specific antibody responses were detected, and only low-magnitude HIV-specific T-cell responses were detected in 8 (53%) of 15 vaccinees. This initial product led to the development of a 4-plasmid multiclade HIV DNA Vaccine Research Center vaccine candidate in which envelope genes expressing Env from clades A, B, and C and a Nef gene from clade B have been added.
Collapse
Affiliation(s)
- Jorge A. Tavel
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Julie E. Martin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Grace G. Kelly
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mary E. Enama
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jean M. Shen
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Phillip L. Gomez
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Charla A. Andrews
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Robert T. Bailer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Judy A. Stein
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Gary J. Nabel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|