1
|
Chen CW, Saubi N, Kilpeläinen A, Joseph-Munné J. Chimeric Human Papillomavirus-16 Virus-like Particles Presenting P18I10 and T20 Peptides from HIV-1 Envelope Induce HPV16 and HIV-1-Specific Humoral and T Cell-Mediated Immunity in BALB/c Mice. Vaccines (Basel) 2022; 11:vaccines11010015. [PMID: 36679860 PMCID: PMC9861546 DOI: 10.3390/vaccines11010015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
In this study, the HIV-1 P18I10 CTL peptide derived from the V3 loop of HIV-1 gp120 and the T20 anti-fusion peptide of HIV-1 gp41 were inserted into the HPV16 L1 capsid protein to construct chimeric HPV:HIV (L1:P18I10 and L1:T20) VLPs by using the mammalian cell expression system. The HPV:HIV VLPs were purified by chromatography. We demonstrated that the insertion of P18I10 or T20 peptides into the DE loop of HPV16 L1 capsid proteins did not affect in vitro stability, self-assembly and morphology of chimeric HPV:HIV VLPs. Importantly, it did not interfere either with the HIV-1 antibody reactivity targeting sequential and conformational P18I10 and T20 peptides presented on chimeric HPV:HIV VLPs or with the induction of HPV16 L1-specific antibodies in vivo. We observed that chimeric L1:P18I10/L1:T20 VLPs vaccines could induce HPV16- but weak HIV-1-specific antibody responses and elicited HPV16- and HIV-1-specific T-cell responses in BALB/c mice. Moreover, could be a potential booster to increase HIV-specific cellular responses in the heterologous immunization after priming with rBCG.HIVA vaccine. This research work would contribute a step towards the development of the novel chimeric HPV:HIV VLP-based vaccine platform for controlling HPV16 and HIV-1 infection, which is urgently needed in developing and industrialized countries.
Collapse
Affiliation(s)
- Chun-Wei Chen
- Department of Biomedical Sciences, University of Barcelona, 08036 Barcelona, Spain
- Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Narcís Saubi
- Respiratory Viruses Unit, Virology Section, Microbiology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Athina Kilpeläinen
- Department of Biomedical Sciences, University of Barcelona, 08036 Barcelona, Spain
- Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Joan Joseph-Munné
- Department of Microbiology, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain
- Correspondence:
| |
Collapse
|
2
|
Marcus H, Thompson E, Zhou Y, Bailey M, Donaldson MM, Stanley DA, Asiedu C, Foulds KE, Roederer M, Moliva JI, Sullivan NJ. Ebola-GP DNA Prime rAd5-GP Boost: Influence of Prime Frequency and Prime/Boost Time Interval on the Immune Response in Non-human Primates. Front Immunol 2021; 12:627688. [PMID: 33790899 PMCID: PMC8006325 DOI: 10.3389/fimmu.2021.627688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/19/2021] [Indexed: 11/13/2022] Open
Abstract
Heterologous prime-boost immunization regimens are a common strategy for many vaccines. DNA prime rAd5-GP boost immunization has been demonstrated to protect non-human primates against a lethal challenge of Ebola virus, a pathogen that causes fatal hemorrhagic disease in humans. This protection correlates with antibody responses and is also associated with IFNγ+ TNFα+ double positive CD8+ T-cells. In this study, we compared single DNA vs. multiple DNA prime immunizations, and short vs. long time intervals between the DNA prime and the rAd5 boost to evaluate the impact of these different prime-boost strategies on vaccine-induced humoral and cellular responses in non-human primates. We demonstrated that DNA/rAd5 prime-boost strategies can be tailored to induce either CD4+ T-cell or CD8+ T-cell dominant responses while maintaining a high magnitude antibody response. Additionally, a single DNA prime immunization generated a stable memory response that could be boosted by rAd5 3 years later. These results suggest DNA/rAd5 prime-boost provides a flexible platform that can be fine-tuned to generate desirable T-cell memory responses.
Collapse
Affiliation(s)
- Hadar Marcus
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Emily Thompson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yan Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michael Bailey
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Mitzi M Donaldson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Daphne A Stanley
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Clement Asiedu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
3
|
Infection of Chinese Rhesus Monkeys with a Subtype C SHIV Resulted in Attenuated In Vivo Viral Replication Despite Successful Animal-to-Animal Serial Passages. Viruses 2021; 13:v13030397. [PMID: 33801437 PMCID: PMC7998229 DOI: 10.3390/v13030397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 01/23/2023] Open
Abstract
Rhesus macaques can be readily infected with chimeric simian-human immunodeficiency viruses (SHIV) as a suitable virus challenge system for testing the efficacy of HIV vaccines. Three Chinese-origin rhesus macaques (ChRM) were inoculated intravenously (IV) with SHIVC109P4 in a rapid serial in vivo passage. SHIV recovered from the peripheral blood of the final ChRM was used to generate a ChRM-adapted virus challenge stock. This stock was titrated for the intrarectal route (IR) in 8 ChRMs using undiluted, 1:10 or 1:100 dilutions, to determine a suitable dose for use in future vaccine efficacy testing via repeated low-dose IR challenges. All 11 ChRMs were successfully infected, reaching similar median peak viraemias at 1–2 weeks post inoculation but undetectable levels by 8 weeks post inoculation. T-cell responses were detected in all animals and Tier 1 neutralizing antibodies (Nab) developed in 10 of 11 infected ChRMs. All ChRMs remained healthy and maintained normal CD4+ T cell counts. Sequence analyses showed >98% amino acid identity between the original inoculum and virus recovered at peak viraemia indicating only minimal changes in the env gene. Thus, while replication is limited over time, our adapted SHIV can be used to test for protection of virus acquisition in ChRMs.
Collapse
|
4
|
Sharan R, Kaushal D. Vaccine strategies for the Mtb/HIV copandemic. NPJ Vaccines 2020; 5:95. [PMID: 33083030 PMCID: PMC7555484 DOI: 10.1038/s41541-020-00245-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
One-third of world’s population is predicted to be infected with tuberculosis (TB). The resurgence of this deadly disease has been inflamed by comorbidity with human immunodeficiency virus (HIV). The risk of TB in people living with HIV (PLWH) is 15–22 times higher than people without HIV. Development of a single vaccine to combat both diseases is an ardent but tenable ambition. Studies have focused on the induction of specific humoral and cellular immune responses against HIV-1 following recombinant BCG (rBCG) expressing HIV-1 antigens. Recent advances in the TB vaccines led to the development of promising candidates such as MTBVAC, the BCG revaccination approach, H4:IC31, H56:IC31, M72/AS01 and more recently, intravenous (IV) BCG. Modification of these vaccine candidates against TB/HIV coinfection could reveal key correlates of protection in a representative animal model. This review discusses the (i) potential TB vaccine candidates that can be exploited for use as a dual vaccine against TB/HIV copandemic (ii) progress made in the realm of TB/HIV dual vaccine candidates in small animal model, NHP model, and human clinical trials (iii) the failures and promising targets for a successful vaccine strategy while delineating the correlates of vaccine-induced protection.
Collapse
Affiliation(s)
- Riti Sharan
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX 78227 USA
| | - Deepak Kaushal
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX 78227 USA
| |
Collapse
|
5
|
Sharan R, Bucşan AN, Ganatra S, Paiardini M, Mohan M, Mehra S, Khader SA, Kaushal D. Chronic Immune Activation in TB/HIV Co-infection. Trends Microbiol 2020; 28:619-632. [PMID: 32417227 PMCID: PMC7390597 DOI: 10.1016/j.tim.2020.03.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/03/2020] [Accepted: 03/25/2020] [Indexed: 12/27/2022]
Abstract
HIV co-infection is the most critical risk factor for the reactivation of latent tuberculosis (TB) infection (LTBI). While CD4+ T cell depletion has been considered the major cause of HIV-induced reactivation of LTBI, recent work in macaques co-infected with Mycobacterium tuberculosis (Mtb)/simian immunodeficiency virus (SIV) suggests that cytopathic effects of SIV resulting in chronic immune activation and dysregulation of T cell homeostasis correlate with reactivation of LTBI. This review builds on compelling data that the reactivation of LTBI during HIV co-infection is likely to be driven by the events of HIV replication and therefore highlights the need to have optimum translational interventions directed at reactivation due to co-infection.
Collapse
Affiliation(s)
- Riti Sharan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Allison N Bucşan
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Shashank Ganatra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Mirko Paiardini
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| |
Collapse
|
6
|
Kilpeläinen A, Saubi N, Guitart N, Moyo N, Wee EG, Ravi K, Hanke T, Joseph J. Priming With Recombinant BCG Expressing Novel HIV-1 Conserved Mosaic Immunogens and Boosting With Recombinant ChAdOx1 Is Safe, Stable, and Elicits HIV-1-Specific T-Cell Responses in BALB/c Mice. Front Immunol 2019; 10:923. [PMID: 31156614 PMCID: PMC6530512 DOI: 10.3389/fimmu.2019.00923] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/10/2019] [Indexed: 11/13/2022] Open
Abstract
BCG is currently the only licensed vaccine against tuberculosis (TB) and confers protection against meningitis and miliary tuberculosis in infants, although pulmonary disease protection in adults is inconsistent. Recently, promising HIV-1 immunogens were developed, such as the T-cell immunogens "tHIVconsvX," designed using functionally conserved protein regions across group M strains, with mosaic immunogens to improve HIV-1 variant match and response breadth. In this study, we constructed an integrative E. coli-mycobacterial shuttle plasmid, p2auxo.HIVconsvXint, expressing the immunogens HIVconsv1&2. This expression vector used an antibiotic resistance-free mechanism for plasmid selection and maintenance. It was first transformed into a glycine auxotrophic E. coli strain and subsequently transformed into a lysine auxotrophic Mycobacterium bovis BCG strain to generate vaccines BCG.HIVconsv12auxo.int and BCG.HIVconsv22auxo.int. The DNA sequence coding for the HIVconsv1&2 immunogens and protein expression were confirmed and working vaccine stocks were genetically and phenotypically characterized. We demonstrated that BCG.HIVconsv1&22auxo.int in combination with ChAdOx1.tHIVconsv5&6 were well tolerated and induced HIV-1-specific T-cell responses in adult BALB/c mice. In addition, we showed that the BCG.HIVconsv1&22auxo.int vaccine strains were stable in vitro after 35 bacterial generations and in vivo 7 weeks after inoculation. The use of integrative expression vectors and novel HIV-1 immunogens are likely to have improved the mycobacterial vaccine stability and specific immunogenicity and may enable the development of a useful vaccine platform for priming protective responses against HIV-1/TB and other prevalent pediatric pathogens shortly following birth.
Collapse
Affiliation(s)
- Athina Kilpeläinen
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| | - Narcís Saubi
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| | - Núria Guitart
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| | - Nathifa Moyo
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | - Edmund G. Wee
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | - Krupa Ravi
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | - Tomáš Hanke
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Joan Joseph
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| |
Collapse
|
7
|
Mahant A, Saubi N, Eto Y, Guitart N, Gatell JM, Hanke T, Joseph J. Preclinical development of BCG.HIVA 2auxo.int, harboring an integrative expression vector, for a HIV-TB Pediatric vaccine. Enhancement of stability and specific HIV-1 T-cell immunity. Hum Vaccin Immunother 2017; 13:1798-1810. [PMID: 28426273 PMCID: PMC5557246 DOI: 10.1080/21645515.2017.1316911] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
One of the critical issues that should be addressed in the development of a BCG-based HIV vaccine is genetic plasmid stability. Therefore, to address this issue we have considered using integrative vectors and the auxotrophic mutant of BCG complemented with a plasmid carrying a wild-type complementing gene. In this study, we have constructed an integrative E. coli-mycobacterial shuttle plasmid, p2auxo.HIVAint, expressing the HIV-1 clade A immunogen HIVA. This shuttle vector uses an antibiotic resistance-free mechanism for plasmid selection and maintenance. It was first transformed into a glycine auxotrophic E. coli strain and subsequently transformed into a lysine auxotrophic Mycobacterium bovis BCG strain to generate the vaccine BCG.HIVA2auxo.int. Presence of the HIVA gene sequence and protein expression was confirmed. We demonstrated that the in vitro stability of the integrative plasmid p2auxo.HIVAint was increased 4-fold, as compared with the BCG strain harboring the episomal plasmid, and was genetically and phenotypically characterized. The BCG.HIVA2auxo.int vaccine in combination with modified vaccinia virus Ankara (MVA).HIVA was found to be safe and induced HIV-1 and Mycobacterium tuberculosis-specific interferon-γ-producing T-cell responses in adult BALB/c mice. We have engineered a more stable and immunogenic BCG-vectored vaccine using the prototype immunogen HIVA. Thus, the use of integrative expression vectors and the antibiotic-free plasmid selection system based on “double” auxotrophic complementation are likely to improve the mycobacterial vaccine stability in vivo and immunogenicity to develop not only recombinant BCG-based vaccines expressing second generation of HIV-1 immunogens but also other major pediatric pathogens to prime protective responses shortly following birth.
Collapse
Affiliation(s)
- Aakash Mahant
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Narcís Saubi
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Yoshiki Eto
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Núria Guitart
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Josep Ma Gatell
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Tomáš Hanke
- b The Jenner Institute , University of Oxford , Oxford , UK
| | - Joan Joseph
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| |
Collapse
|
8
|
Chege GK, Burgers WA, Müller TL, Gray CM, Shephard EG, Barnett SW, Ferrari G, Montefiori D, Williamson C, Williamson AL. DNA-MVA-protein vaccination of rhesus macaques induces HIV-specific immunity in mucosal-associated lymph nodes and functional antibodies. Vaccine 2017; 35:929-937. [PMID: 28069361 PMCID: PMC5287223 DOI: 10.1016/j.vaccine.2016.12.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/06/2016] [Accepted: 12/23/2016] [Indexed: 11/30/2022]
Abstract
Successful future HIV vaccines are expected to generate an effective cellular and humoral response against the virus in both the peripheral blood and mucosal compartments. We previously reported the development of DNA-C and MVA-C vaccines based on HIV-1 subtype C and demonstrated their immunogenicity when given in a DNA prime-MVA boost combination in a nonhuman primate model. In the current study, rhesus macaques previously vaccinated with a DNA-C and MVA-C vaccine regimen were re-vaccinated 3.5 years later with MVA-C followed by a protein vaccine based on HIV-1 subtype C envelope formulated with MF59 adjuvant (gp140Env/MF59), and finally a concurrent boost with both vaccines. A single MVA-C re-vaccination elicited T cell responses in all animals similar to previous peak responses, with 4/7 demonstrating responses >1000 SFU/106 PBMC. In contrast to an Env/MF59-only vaccine, concurrent boosting with MVA-C and Env/MF59 induced HIV-specific cellular responses in multiple mucosal associated lymph nodes in 6/7 animals, with high magnitude responses in some animals. Both vaccine regimens induced high titer Env-specific antibodies with ADCC activity, as well as neutralization of Tier 1 viruses and modest Tier 2 neutralization. These data demonstrate the feasibility of inducing HIV-specific immunity in the blood and mucosal sites of viral entry by means of DNA and poxvirus-vectored vaccines, in combination with a HIV envelope-based protein vaccine.
Collapse
Affiliation(s)
- Gerald K Chege
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Wendy A Burgers
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Tracey L Müller
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Clive M Gray
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Enid G Shephard
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; South African Medical Research Council, Cape Town, South Africa
| | | | | | | | - Carolyn Williamson
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Anna-Lise Williamson
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa.
| |
Collapse
|
9
|
Williamson AL, Rybicki EP. Justification for the inclusion of Gag in HIV vaccine candidates. Expert Rev Vaccines 2015; 15:585-98. [PMID: 26645951 DOI: 10.1586/14760584.2016.1129904] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
It is widely accepted that effective human immunodeficiency virus (HIV) vaccines need to elicit a range of responses, including neutralising antibodies and T-cells. In natural HIV infections, immune responses to Gag are associated with lower viral load in infected individuals, and these responses can be measured against infected cells before the replication of HIV. Priming immune responses to Gag with DNA or recombinant Bacillus Calmette-Guérin (BCG) vaccines, and boosting with Gag virus-like particles as subunit vaccines or Gag produced in vivo by other vaccine vectors, elicits high-magnitude, broad polyfunctional responses, with memory T-cell responses appropriate for virus control. This review provides justification for the inclusion of HIV Gag in vaccine regimens, either as a transgene expressing protein that may assemble to form budded particles, or as purified virus-like particles. Possible benefits would include early control via CD8(+) T-cells at the site of infection, control of spread from the entry portal, and control of viraemia if infection is established.
Collapse
Affiliation(s)
- Anna-Lise Williamson
- a Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town , South Africa.,b National Health Laboratory Service, Groote Schuur Hospital, Cape Town and Department of Pathology , University of Cape Town , Cape Town , South Africa
| | - Edward P Rybicki
- a Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town , South Africa.,c Biopharming Research Unit, Department of Molecular and Cell Biology , University of Cape Town , Cape Town , South Africa
| |
Collapse
|
10
|
Zheng YQ, Naguib YW, Dong Y, Shi YC, Bou S, Cui Z. Applications of bacillus Calmette–Guerin and recombinant bacillus Calmette–Guerin in vaccine development and tumor immunotherapy. Expert Rev Vaccines 2015. [DOI: 10.1586/14760584.2015.1068124] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yuan-qiang Zheng
- 1Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
| | - Youssef W Naguib
- 2Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yixuan Dong
- 2Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yan-chun Shi
- 1Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
| | - Shorgan Bou
- 3National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, China
| | - Zhengrong Cui
- 1Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
- 2Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
11
|
Abstract
Plant-made or "biofarmed" viral vaccines are some of the earliest products of the technology of plant molecular farming, and remain some of the brightest prospects for the success of this field. Proofs of principle and of efficacy exist for many candidate viral veterinary vaccines; the use of plant-made viral antigens and of monoclonal antibodies for therapy of animal and even human viral disease is also well established. This review explores some of the more prominent recent advances in the biofarming of viral vaccines and therapies, including the recent use of ZMapp for Ebolavirus infection, and explores some possible future applications of the technology.
Collapse
Affiliation(s)
- Edward P Rybicki
- Biopharming Research Unit, Department of Molecular & Cell Biology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Private Bag X3, Rondebosch, 7701, Cape Town, South Africa.
| |
Collapse
|
12
|
Saubi N, Gea-Mallorquí E, Ferrer P, Hurtado C, Sánchez-Úbeda S, Eto Y, Gatell JM, Hanke T, Joseph J. Engineering new mycobacterial vaccine design for HIV-TB pediatric vaccine vectored by lysine auxotroph of BCG. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14017. [PMID: 26015961 PMCID: PMC4362382 DOI: 10.1038/mtm.2014.17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/26/2014] [Indexed: 02/05/2023]
Abstract
In this study, we have engineered a new mycobacterial vaccine design by using an antibiotic-free plasmid selection system. We assembled a novel Escherichia coli (E. coli)–mycobacterial shuttle plasmid p2auxo.HIVA, expressing the HIV-1 clade A immunogen HIVA. This shuttle vector employs an antibiotic resistance-free mechanism for plasmid selection and maintenance based on glycine complementation in E. coli and lysine complementation in mycobacteria. This plasmid was first transformed into glycine auxotroph of E. coli strain and subsequently transformed into lysine auxotroph of Mycobacterium bovis BCG strain to generate vaccine BCG.HIVA2auxo. We demonstrated that the episomal plasmid p2auxo.HIVA was stable in vivo over a 7-week period and genetically and phenotypically characterized the BCG.HIVA2auxo vaccine strain. The BCG.HIVA2auxo vaccine in combination with modified vaccinia virus Ankara (MVA). HIVA was safe and induced HIV-1 and Mycobacterium tuberculosis-specific interferon-γ-producing T-cell responses in adult BALB/c mice. Polyfunctional HIV-1-specific CD8+ T cells, which produce interferon-γ and tumor necrosis factor-α and express the degranulation marker CD107a, were induced. Thus, we engineered a novel, safer, good laboratory practice–compatible BCG-vectored vaccine using prototype immunogen HIVA. This antibiotic-free plasmid selection system based on “double” auxotrophic complementation might be a new mycobacterial vaccine platform to develop not only recombinant BCG-based vaccines expressing second generation of HIV-1 immunogens but also other major pediatric pathogens to prime protective response soon after birth.
Collapse
Affiliation(s)
- Narcís Saubi
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Ester Gea-Mallorquí
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Pau Ferrer
- Department of Chemical Engineering, Group of Bioprocess Engineering and Applied Biocatalysis, School of Engineering, Autonomous University of Barcelona , Barcelona, Catalonia, Spain
| | - Carmen Hurtado
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Sara Sánchez-Úbeda
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Yoshiki Eto
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Josep M Gatell
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford , Oxford, UK ; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford, UK
| | - Joan Joseph
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| |
Collapse
|
13
|
Scotti N, Rybicki EP. Virus-like particles produced in plants as potential vaccines. Expert Rev Vaccines 2014; 12:211-24. [DOI: 10.1586/erv.12.147] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
14
|
Robust immunity to an auxotrophic Mycobacterium bovis BCG-VLP prime-boost HIV vaccine candidate in a nonhuman primate model. J Virol 2013; 87:5151-60. [PMID: 23449790 DOI: 10.1128/jvi.03178-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We previously reported that a recombinant pantothenate auxotroph of Mycobacterium bovis BCG expressing human immunodeficiency virus type 1 (HIV-1) subtype C Gag (rBCGpan-Gag) efficiently primes the mouse immune system for a boost with a recombinant modified vaccinia virus Ankara (rMVA) vaccine. In this study, we further evaluated the immunogenicity of rBCGpan-Gag in a nonhuman primate model. Two groups of chacma baboons were primed or mock primed twice with either rBCGpan-Gag or a control BCG. Both groups were boosted with HIV-1 Pr55(gag) virus-like particles (Gag VLPs). The magnitude and breadth of HIV-specific cellular responses were measured using a gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISPOT) assay, and the cytokine profiles and memory phenotypes of T cells were evaluated by polychromatic flow cytometry. Gag-specific responses were detected in all animals after the second inoculation with rBCGpan-Gag. Boosting with Gag VLPs significantly increased the magnitude and breadth of the responses in the baboons that were primed with rBCGpan-Gag. These responses targeted an average of 12 Gag peptides per animal, compared to an average of 3 peptides per animal for the mock-primed controls. Robust responses of Gag-specific polyfunctional T cells capable of simultaneously producing IFN-γ, tumor necrosis alpha (TNF-α), and interleukin-2 (IL-2) were detected in the rBCGpan-Gag-primed animals. Gag-specific memory T cells were skewed toward a central memory phenotype in both CD4(+) and CD8(+) T cell populations. These data show that the rBCGpan-Gag prime and Gag VLP boost vaccine regimen is highly immunogenic, inducing a broad and polyfunctional central memory T cell response. This report further indicates the feasibility of developing a BCG-based HIV vaccine that is safe for childhood HIV immunization.
Collapse
|
15
|
Lynch A, Meyers AE, Williamson AL, Rybicki EP. Stability studies of HIV-1 Pr55gag virus-like particles made in insect cells after storage in various formulation media. Virol J 2012; 9:210. [PMID: 22988963 PMCID: PMC3502365 DOI: 10.1186/1743-422x-9-210] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 09/17/2012] [Indexed: 11/23/2022] Open
Abstract
Background HIV-1 Pr55gag virus-like particles (VLPs) expressed by baculovirus in insect cells are considered to be a very promising HIV-1 vaccine candidate, as they have been shown to elicit broad cellular immune responses when tested in animals, particularly when used as a boost to DNA or BCG vaccines. However, it is important for the VLPs to retain their structure for them to be fully functional and effective. The medium in which the VLPs are formulated and the temperature at which they are stored are two important factors affecting their stability. Findings We describe the screening of 3 different readily available formulation media (sorbitol, sucrose and trehalose) for their ability to stabilise HIV-1 Pr55gag VLPs during prolonged storage. Transmission electron microscopy (TEM) was done on VLPs stored at two different concentrations of the media at three different temperatures (4°C, –20°C and −70°C) over different time periods, and the appearance of the VLPs was compared. VLPs stored in 15% trehalose at −70°C retained their original appearance the most effectively over a period of 12 months. VLPs stored in 5% trehalose, sorbitol or sucrose were not all intact even after 1 month storage at the temperatures tested. In addition, we showed that VLPs stored under these conditions were able to be frozen and re-thawed twice before showing changes in their appearance. Conclusions Although the inclusion of other analytical tools are essential to validate these preliminary findings, storage in 15% trehalose at −70°C for 12 months is most effective in retaining VLP stability.
Collapse
Affiliation(s)
- Alisson Lynch
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, University Ave, Rondebosch, 7701, South Africa
| | | | | | | |
Collapse
|
16
|
Saubi N, Mbewe-Mvula A, Gea-Mallorqui E, Rosario M, Gatell JM, Hanke T, Joseph J. Pre-clinical development of BCG.HIVA(CAT), an antibiotic-free selection strain, for HIV-TB pediatric vaccine vectored by lysine auxotroph of BCG. PLoS One 2012; 7:e42559. [PMID: 22927933 PMCID: PMC3424164 DOI: 10.1371/journal.pone.0042559] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 07/09/2012] [Indexed: 01/13/2023] Open
Abstract
In the past, we proposed to develop a heterologous recombinant BCG prime-recombinant modified vaccinia virus Ankara (MVA) boost dual pediatric vaccine platform against transmission of breast milk HIV-1 and Mycobacterium tuberculosis (Mtb). In this study, we assembled an E. coli-mycobacterial shuttle plasmid pJH222.HIVACAT expressing HIV-1 clade A immunogen HIVA. This shuttle vector employs an antibiotic resistance-free mechanism based on Operator-Repressor Titration (ORT) system for plasmid selection and maintenance in E. coli and lysine complementation in mycobacteria. This shuttle plasmid was electroporated into parental lysine auxotroph (safer) strain of BCG to generate vaccine BCG.HIVACAT. All procedures complied with Good Laboratory Practices (GLPs). We demonstrated that the episomal plasmid pJH222.HIVACAT was stable in vivo over a 20-week period, and genetically and phenotypically characterized the BCG.HIVACAT vaccine strain. The BCG.HIVACAT vaccine in combination with MVA.HIVA induced HIV-1- and Mtb-specific interferon γ-producing T-cell responses in newborn and adult BALB/c mice. On the other hand, when adult mice were primed with BCG.HIVACAT and boosted with MVA.HIVA.85A, HIV-1-specific CD8+ T-cells producing IFN-γ, TNF-α, IL-2 and CD107a were induced. To assess the biosafety profile of BCG.HIVACAT-MVA.HIVA regimen, body mass loss of newborn mice was monitored regularly throughout the vaccination experiment and no difference was observed between the vaccinated and naïve groups of animals. Thus, we demonstrated T-cell immunogenicity of a novel, safer, GLP-compatible BCG-vectored vaccine using prototype immunogen HIVA. Second generation immunogens derived from HIV-1 as well as other major pediatric pathogens can be constructed in a similar fashion to prime protective responses soon after birth.
Collapse
Affiliation(s)
- Narcís Saubi
- AIDS Research Group, Hospital Clinic/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Ester Gea-Mallorqui
- AIDS Research Group, Hospital Clinic/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Maximillian Rosario
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Josep Maria Gatell
- AIDS Research Group, Hospital Clinic/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Joan Joseph
- AIDS Research Group, Hospital Clinic/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
17
|
Chapman R, Shephard E, Stutz H, Douglass N, Sambandamurthy V, Garcia I, Ryffel B, Jacobs W, Williamson AL. Priming with a recombinant pantothenate auxotroph of Mycobacterium bovis BCG and boosting with MVA elicits HIV-1 Gag specific CD8+ T cells. PLoS One 2012; 7:e32769. [PMID: 22479338 PMCID: PMC3315557 DOI: 10.1371/journal.pone.0032769] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 01/30/2012] [Indexed: 12/03/2022] Open
Abstract
A safe and effective HIV vaccine is required to significantly reduce the number of people becoming infected with HIV each year. In this study wild type Mycobacterium bovis BCG Pasteur and an attenuated pantothenate auxotroph strain (BCGΔpanCD) that is safe in SCID mice, have been compared as vaccine vectors for HIV-1 subtype C Gag. Genetically stable vaccines BCG[pHS400] (BCG-Gag) and BCGΔpanCD[pHS400] (BCGpan-Gag) were generated using the Pasteur strain of BCG, and a panothenate auxotroph of Pasteur respectively. Stability was achieved by the use of a codon optimised gag gene and deletion of the hsp60-lysA promoter-gene cassette from the episomal vector pCB119. In this vector expression of gag is driven by the mtrA promoter and the Gag protein is fused to the Mycobacterium tuberculosis 19 kDa signal sequence. Both BCG-Gag and BCGpan-Gag primed the immune system of BALB/c mice for a boost with a recombinant modified vaccinia virus Ankara expressing Gag (MVA-Gag). After the boost high frequencies of predominantly Gag-specific CD8(+) T cells were detected when BCGpan-Gag was the prime in contrast to induction of predominantly Gag-specific CD4(+) T cells when priming with BCG-Gag. The differing Gag-specific T-cell phenotype elicited by the prime-boost regimens may be related to the reduced inflammation observed with the pantothenate auxotroph strain compared to the parent strain. These features make BCGpan-Gag a more desirable HIV vaccine candidate than BCG-Gag. Although no Gag-specific cells could be detected after vaccination of BALB/c mice with either recombinant BCG vaccine alone, BCGpan-Gag protected mice against a surrogate vaccinia virus challenge.
Collapse
Affiliation(s)
- Rosamund Chapman
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Clinical Laboratory Science, University of Cape Town, Cape Town, South Africa
| | - Enid Shephard
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Medical Research Council, Cape Town, South Africa
| | - Helen Stutz
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Clinical Laboratory Science, University of Cape Town, Cape Town, South Africa
| | - Nicola Douglass
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Clinical Laboratory Science, University of Cape Town, Cape Town, South Africa
| | | | - Irene Garcia
- Department of Pathology and Immunology, Centre Médical Universitaire, Hôpitaux Universitaires de Genève, University of Geneva, Geneva, Switzerland
| | - Bernhard Ryffel
- University of Orleans and Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Molecular Immunology and Embryology, Orleans, France
| | - William Jacobs
- Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Clinical Laboratory Science, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| |
Collapse
|
18
|
Bridge SH, Sharpe SA, Dennis MJ, Dowall SD, Getty B, Anson DS, Skinner MA, Stewart JP, Blanchard TJ. Heterologous prime-boost-boost immunisation of Chinese cynomolgus macaques using DNA and recombinant poxvirus vectors expressing HIV-1 virus-like particles. Virol J 2011; 8:429. [PMID: 21899739 PMCID: PMC3177910 DOI: 10.1186/1743-422x-8-429] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 09/07/2011] [Indexed: 01/13/2023] Open
Abstract
Background There is renewed interest in the development of poxvirus vector-based HIV vaccines due to the protective effect observed with repeated recombinant canarypox priming with gp120 boosting in the recent Thai placebo-controlled trial. This study sought to investigate whether a heterologous prime-boost-boost vaccine regimen in Chinese cynomolgus macaques with a DNA vaccine and recombinant poxviral vectors expressing HIV virus-like particles bearing envelopes derived from the most prevalent clades circulating in sub-Saharan Africa, focused the antibody response to shared neutralising epitopes. Methods Three Chinese cynomolgus macaques were immunised via intramuscular injections using a regimen composed of a prime with two DNA vaccines expressing clade A Env/clade B Gag followed by boosting with recombinant fowlpox virus expressing HIV-1 clade D Gag, Env and cholera toxin B subunit followed by the final boost with recombinant modified vaccinia virus Ankara expressing HIV-1 clade C Env, Gag and human complement protein C3d. We measured the macaque serum antibody responses by ELISA, enumerated T cell responses by IFN-γ ELISpot and assessed seroneutralisation of HIV-1 using the TZM-bl β-galactosidase assay with primary isolates of HIV-1. Results This study shows that large and complex synthetic DNA sequences can be successfully cloned in a single step into two poxvirus vectors: MVA and FPV and the recombinant poxviruses could be grown to high titres. The vaccine candidates showed appropriate expression of recombinant proteins with the formation of authentic HIV virus-like particles seen on transmission electron microscopy. In addition the b12 epitope was shown to be held in common by the vaccine candidates using confocal immunofluorescent microscopy. The vaccine candidates were safely administered to Chinese cynomolgus macaques which elicited modest T cell responses at the end of the study but only one out of the three macaques elicited an HIV-specific antibody response. However, the antibodies did not neutralise primary isolates of HIV-1 or the V3-sensitive isolate SF162 using the TZM-bl β-galactosidase assay. Conclusions MVA and FP9 are ideal replication-deficient viral vectors for HIV-1 vaccines due to their excellent safety profile for use in humans. This study shows this novel prime-boost-boost regimen was poorly immunogenic in Chinese cynomolgus macaques.
Collapse
Affiliation(s)
- Simon H Bridge
- Clinical Research Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hopkins R, Bridgeman A, Joseph J, Gilbert SC, McShane H, Hanke T. Dual neonate vaccine platform against HIV-1 and M. tuberculosis. PLoS One 2011; 6:e20067. [PMID: 21603645 PMCID: PMC3094449 DOI: 10.1371/journal.pone.0020067] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 04/20/2011] [Indexed: 01/05/2023] Open
Abstract
Acquired immunodeficiency syndrome and tuberculosis (TB) are two of the
world's most devastating diseases. The first vaccine the majority of
infants born in Africa receive is Mycobacterium bovis bacillus
Calmette-Guérin (BCG) as a prevention against TB. BCG protects against
disseminated disease in the first 10 years of life, but provides a variable
protection against pulmonary TB and enhancing boost delivered by recombinant
modified vaccinia virus Ankara (rMVA) expressing antigen 85A (Ag85A) of
M. tuberculosis is currently in phase IIb evaluation in
African neonates. If the newborn's mother is positive for human
immunodeficiency virus type 1 (HIV-1), the baby is at high risk of acquiring
HIV-1 through breastfeeding. We suggested that a vaccination consisting of
recombinant BCG expressing HIV-1 immunogen administered at birth followed by a
boost with rMVA sharing the same immunogen could serve as a strategy for
prevention of mother-to-child transmission of HIV-1 and rMVA expressing an
African HIV-1-derived immunogen HIVA is currently in phase I trials in African
neonates. Here, we aim to develop a dual neonate vaccine platform against HIV-1
and TB consisting of BCG.HIVA administered at birth followed by a boost with
MVA.HIVA.85A. Thus, mMVA.HIVA.85A and sMVA.HIVA.85A vaccines were constructed,
in which the transgene transcription is driven by either modified H5 or short
synthetic promoters, respectively, and tested for immunogenicity alone and in
combination with BCG.HIVA222. mMVA.HIVA.85A was produced markerless
and thus suitable for clinical manufacture. While sMVA.HIVA.85A expressed higher
levels of the immunogens, it was less immunogenic than mMVA.HIVA.85A in BALB/c
mice. A BCG.HIVA222–mMVA.HIVA.85A prime-boost regimen induced
robust T cell responses to both HIV-1 and M. tuberculosis.
Therefore, proof-of-principle for a dual anti-HIV-1/M.
tuberculosis infant vaccine platform is established. Induction of
immune responses against these pathogens soon after birth is highly desirable
and may provide a basis for lifetime protection maintained by boosts later in
life.
Collapse
Affiliation(s)
- Richard Hopkins
- MRC Human Immunology Unit, Weatherall
Institute of Molecular Medicine, University of Oxford, Oxford, United
Kingdom
| | - Anne Bridgeman
- MRC Human Immunology Unit, Weatherall
Institute of Molecular Medicine, University of Oxford, Oxford, United
Kingdom
| | - Joan Joseph
- AIDS Research Unit, Hospital
Clínic/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona,
Barcelona, Spain
| | - Sarah C. Gilbert
- The Jenner Institute, University of Oxford,
Oxford, United Kingdom
| | - Helen McShane
- The Jenner Institute, University of Oxford,
Oxford, United Kingdom
| | - Tomáš Hanke
- MRC Human Immunology Unit, Weatherall
Institute of Molecular Medicine, University of Oxford, Oxford, United
Kingdom
- The Jenner Institute, University of Oxford,
Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Pillay S, Shephard EG, Meyers AE, Williamson AL, Rybicki EP. HIV-1 sub-type C chimaeric VLPs boost cellular immune responses in mice. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2010; 8:7. [PMID: 21087527 PMCID: PMC2996383 DOI: 10.1186/1476-8518-8-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 11/19/2010] [Indexed: 11/30/2022]
Abstract
Several approaches have been explored to eradicate HIV; however, a multigene vaccine appears to be the best option, given their proven potential to elicit broad, effective responses in animal models. The Pr55Gag protein is an excellent vaccine candidate in its own right, given that it can assemble into large, enveloped, virus-like particles (VLPs) which are highly immunogenic, and can moreover be used as a scaffold for the presentation of other large non-structural HIV antigens. In this study, we evaluated the potential of two novel chimaeric HIV-1 Pr55Gag-based VLP constructs - C-terminal fusions with reverse transcriptase and a Tat::Nef fusion protein, designated GagRT and GagTN respectively - to enhance a cellular response in mice when used as boost components in two types of heterologous prime-boost vaccine strategies. A vaccine regimen consisting of a DNA prime and chimaeric HIV-1 VLP boosts in mice induced strong, broad cellular immune responses at an optimum dose of 100 ng VLPs. The enhanced cellular responses induced by the DNA prime-VLP boost were two- to three-fold greater than two DNA vaccinations. Moreover, a mixture of GagRT and GagTN VLPs also boosted antigen-specific CD8+ and CD4+ T-cell responses, while VLP vaccinations only induced predominantly robust Gag CD4+ T-cell responses. The results demonstrate the promising potential of these chimaeric VLPs as vaccine candidates against HIV-1.
Collapse
Affiliation(s)
- Sirika Pillay
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, University Ave, Rondebosch 7701, South Africa.
| | | | | | | | | |
Collapse
|
21
|
Rybicki EP. Plant-made vaccines for humans and animals. PLANT BIOTECHNOLOGY JOURNAL 2010; 8:620-37. [PMID: 20233333 PMCID: PMC7167690 DOI: 10.1111/j.1467-7652.2010.00507.x] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 11/30/2009] [Accepted: 12/02/2009] [Indexed: 05/17/2023]
Abstract
The concept of using plants to produce high-value pharmaceuticals such as vaccines is 20 years old this year and is only now on the brink of realisation as an established technology. The original reliance on transgenic plants has largely given way to transient expression; proofs of concept for human and animal vaccines and of efficacy for animal vaccines have been established; several plant-produced vaccines have been through Phase I clinical trials in humans and more are scheduled; regulatory requirements are more clear than ever, and more facilities exist for manufacture of clinic-grade materials. The original concept of cheap edible vaccines has given way to a realisation that formulated products are required, which may well be injectable. The technology has proven its worth as a means of cheap, easily scalable production of materials: it now needs to find its niche in competition with established technologies. The realised achievements in the field as well as promising new developments will be reviewed, such as rapid-response vaccines for emerging viruses with pandemic potential and bioterror agents.
Collapse
Affiliation(s)
- Edward P Rybicki
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, South Africa. ed.rybicki@ uct.ac.za
| |
Collapse
|
22
|
Rosario M, Hopkins R, Fulkerson J, Borthwick N, Quigley MF, Joseph J, Douek DC, Greenaway HY, Venturi V, Gostick E, Price DA, Both GW, Sadoff JC, Hanke T. Novel recombinant Mycobacterium bovis BCG, ovine atadenovirus, and modified vaccinia virus Ankara vaccines combine to induce robust human immunodeficiency virus-specific CD4 and CD8 T-cell responses in rhesus macaques. J Virol 2010; 84:5898-908. [PMID: 20375158 PMCID: PMC2876636 DOI: 10.1128/jvi.02607-09] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 03/30/2010] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium bovis bacillus Calmette-Guérin (BCG), which elicits a degree of protective immunity against tuberculosis, is the most widely used vaccine in the world. Due to its persistence and immunogenicity, BCG has been proposed as a vector for vaccines against other infections, including HIV-1. BCG has a very good safety record, although it can cause disseminated disease in immunocompromised individuals. Here, we constructed a recombinant BCG vector expressing HIV-1 clade A-derived immunogen HIVA using the recently described safer and more immunogenic BCG strain AERAS-401 as the parental mycobacterium. Using routine ex vivo T-cell assays, BCG.HIVA(401) as a stand-alone vaccine induced undetectable and weak CD8 T-cell responses in BALB/c mice and rhesus macaques, respectively. However, when BCG.HIVA(401) was used as a priming component in heterologous vaccination regimens together with recombinant modified vaccinia virus Ankara-vectored MVA.HIVA and ovine atadenovirus-vectored OAdV.HIVA vaccines, robust HIV-1-specific T-cell responses were elicited. These high-frequency T-cell responses were broadly directed and capable of proliferation in response to recall antigen. Furthermore, multiple antigen-specific T-cell clonotypes were efficiently recruited into the memory pool. These desirable features are thought to be associated with good control of HIV-1 infection. In addition, strong and persistent T-cell responses specific for the BCG-derived purified protein derivative (PPD) antigen were induced. This work is the first demonstration of immunogenicity for two novel vaccine vectors and the corresponding candidate HIV-1 vaccines BCG.HIVA(401) and OAdV.HIVA in nonhuman primates. These results strongly support their further exploration.
Collapse
Affiliation(s)
- Maximillian Rosario
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Richard Hopkins
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - John Fulkerson
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Nicola Borthwick
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Máire F. Quigley
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Joan Joseph
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Daniel C. Douek
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Hui Yee Greenaway
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Vanessa Venturi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Emma Gostick
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - David A. Price
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Gerald W. Both
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Jerald C. Sadoff
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| | - Tomáš Hanke
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom, Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, Maryland 20850, Vaccine Research Centre, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, Catalan HIV Vaccine Research and Development Center, AIDS Research Unit, Infectious Diseases Department, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, School of Medicine, University of Barcelona, 170 08036 Barcelona, Spain, Computational Biology Unit, Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia, Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom, Biotech Equity Partners Pty., Ltd., Riverside Life Sciences Building, 11 Julius Ave., North Ryde, New South Wales 2113, Australia
| |
Collapse
|
23
|
Brown SA, Surman SL, Sealy R, Jones BG, Slobod KS, Branum K, Lockey TD, Howlett N, Freiden P, Flynn P, Hurwitz JL. Heterologous Prime-Boost HIV-1 Vaccination Regimens in Pre-Clinical and Clinical Trials. Viruses 2010; 2:435-467. [PMID: 20407589 PMCID: PMC2855973 DOI: 10.3390/v2020435] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/12/2010] [Accepted: 01/22/2010] [Indexed: 12/21/2022] Open
Abstract
Currently, there are more than 30 million people infected with HIV-1 and thousands more are infected each day. Vaccination is the single most effective mechanism for prevention of viral disease, and after more than 25 years of research, one vaccine has shown somewhat encouraging results in an advanced clinical efficacy trial. A modified intent-to-treat analysis of trial results showed that infection was approximately 30% lower in the vaccine group compared to the placebo group. The vaccine was administered using a heterologous prime-boost regimen in which both target antigens and delivery vehicles were changed during the course of inoculations. Here we examine the complexity of heterologous prime-boost immunizations. We show that the use of different delivery vehicles in prime and boost inoculations can help to avert the inhibitory effects caused by vector-specific immune responses. We also show that the introduction of new antigens into boost inoculations can be advantageous, demonstrating that the effect of `original antigenic sin' is not absolute. Pre-clinical and clinical studies are reviewed, including our own work with a three-vector vaccination regimen using recombinant DNA, virus (Sendai virus or vaccinia virus) and protein. Promising preliminary results suggest that the heterologous prime-boost strategy may possibly provide a foundation for the future prevention of HIV-1 infections in humans.
Collapse
Affiliation(s)
- Scott A. Brown
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (S.A.B.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Sherri L. Surman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Robert Sealy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Bart G. Jones
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Karen S. Slobod
- Early Development, Novartis Vaccines and Diagnostics, 350 Mass Ave. Cambridge, MA 02139, USA; E-Mail: (K.S.S.)
| | - Kristen Branum
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Timothy D. Lockey
- Department of Therapeutics, Production and Quality, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (T.D.L.)
| | - Nanna Howlett
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Pamela Freiden
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Patricia Flynn
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
- Department of Pediatrics, University of Tennessee, Memphis, TN 38163, USA
| | - Julia L. Hurwitz
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (S.A.B.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
- Department of Pathology, University of Tennessee, Memphis, TN 38163, USA
| |
Collapse
|
24
|
Challenges and perspectives in vaccination against leishmaniasis. Parasitol Int 2009; 58:319-24. [DOI: 10.1016/j.parint.2009.07.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 07/28/2009] [Accepted: 07/31/2009] [Indexed: 11/24/2022]
|