1
|
Rosen BC, Sawatzki K, Ricciardi MJ, Smith E, Golez I, Mauter JT, Pedreño-López N, Yrizarry-Medina A, Weisgrau KL, Vosler LJ, Voigt TB, Louw JJ, Tisoncik-Go J, Whitmore LS, Panayiotou C, Ghosh N, Furlott JR, Parks CL, Desrosiers RC, Lifson JD, Rakasz EG, Watkins DI, Gale M. Acute-phase innate immune responses in SIVmac239-infected Mamu-B*08+ Indian rhesus macaques may contribute to the establishment of elite control. Front Immunol 2024; 15:1478063. [PMID: 39502699 PMCID: PMC11534762 DOI: 10.3389/fimmu.2024.1478063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Spontaneous control of chronic-phase HIV/SIV viremia is often associated with the expression of specific MHC class I allotypes. HIV/SIV-specific CD8+ cytotoxic T lymphocytes (CTLs) restricted by these MHC class I allotypes appear to be critical for viremic control. Establishment of the elite controller (EC) phenotype is predictable in SIVmac239-infected Indian rhesus macaques (RMs), with approximately 50% of Mamu-B*08+ RMs and 20% of Mamu-B*17+ RMs becoming ECs. Despite extensive characterization of EC-associated CTLs in HIV/SIV-infected individuals, the precise mechanistic basis of elite control remains unknown. Because EC and non-EC viral load trajectories begin diverging by day 14 post-infection, we hypothesized that hyperacute innate immune responses may contribute to viremic control. Methods To gain insight into the immunological factors involved in the determination of EC status, we vaccinated 16 Mamu-B*08+ RMs with Vif and Nef to elicit EC-associated CTLs, then subjected these 16 vaccinees and an additional 16 unvaccinated Mamu-B*08+ controls to repeated intrarectal SIVmac239 challenges. We then performed whole-blood transcriptomic analysis of all 32 SIVmac239-infected Mamu-B*08+ RMs and eight SIVmac239-infected Mamu-B*08 - RMs during the first 14 days of infection. Results Vaccination did not provide protection against acquisition, but peak and setpoint viremia were significantly lower in vaccinees relative to controls. We did not identify any meaningful correlations between vaccine-induced CTL parameters and SIVmac239 acquisition rate or chronic-phase viral loads. Ultimately, 13 of 16 vaccinees (81%) and 7 of 16 controls (44%) became ECs (viremia ≤ 10,000 vRNA copies/mL plasma for ≥ 4 weeks). We identified subsets of immunomodulatory genes differentially expressed (DE) between RM groupings based on vaccination status, EC status, and MHC class I genotype. These DE genes function in multiple innate immune processes, including the complement system, cytokine/chemokine signaling, pattern recognition receptors, and interferon-mediated responses. Discussion A striking difference in the kinetics of differential gene expression among our RM groups suggests that Mamu-B*08-associated elite control is characterized by a robust, rapid innate immune response that quickly resolves. These findings indicate that, despite the association between MHC class I genotype and elite control, innate immune factors in hyperacute SIV infection preceding CTL response development may facilitate the establishment of the EC phenotype.
Collapse
Affiliation(s)
- Brandon C. Rosen
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Kaitlin Sawatzki
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, United States
| | - Michael J. Ricciardi
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Elise Smith
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, United States
| | - Inah Golez
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, United States
| | - Jack T. Mauter
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Núria Pedreño-López
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Aaron Yrizarry-Medina
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Kim L. Weisgrau
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Logan J. Vosler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Thomas B. Voigt
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Johan J. Louw
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Jennifer Tisoncik-Go
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, United States
| | - Leanne S. Whitmore
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, United States
| | - Christakis Panayiotou
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Noor Ghosh
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Jessica R. Furlott
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Ronald C. Desrosiers
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - David I. Watkins
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
2
|
Kaur A, Vaccari M. Exploring HIV Vaccine Progress in the Pre-Clinical and Clinical Setting: From History to Future Prospects. Viruses 2024; 16:368. [PMID: 38543734 PMCID: PMC10974975 DOI: 10.3390/v16030368] [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: 01/09/2024] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 04/01/2024] Open
Abstract
The human immunodeficiency virus (HIV) continues to pose a significant global health challenge, with millions of people affected and new cases emerging each year. While various treatment and prevention methods exist, including antiretroviral therapy and non-vaccine approaches, developing an effective vaccine remains the most crucial and cost-effective solution to combating the HIV epidemic. Despite significant advancements in HIV research, the HIV vaccine field has faced numerous challenges, and only one clinical trial has demonstrated a modest level of efficacy. This review delves into the history of HIV vaccines and the current efforts in HIV prevention, emphasizing pre-clinical vaccine development using the non-human primate model (NHP) of HIV infection. NHP models offer valuable insights into potential preventive strategies for combating HIV, and they play a vital role in informing and guiding the development of novel vaccine candidates before they can proceed to human clinical trials.
Collapse
Affiliation(s)
- Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Monica Vaccari
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
3
|
Ishii H, Terahara K, Nomura T, Okazaki M, Yamamoto H, Shu T, Sakawaki H, Miura T, Watkins DI, Matano T. Env-independent protection of intrarectal SIV challenge by vaccine induction of Gag/Vif-specific CD8+ T cells but not CD4+ T cells. Mol Ther 2022; 30:2048-2057. [PMID: 35231604 PMCID: PMC9092394 DOI: 10.1016/j.ymthe.2022.02.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/20/2022] [Accepted: 02/23/2022] [Indexed: 10/19/2022] Open
Abstract
Effective T cell induction is an important strategy in HIV-vaccine development. However, it has been indicated that vaccine-induced HIV-specific CD4+ T cells, the preferential targets of HIV infection, might increase viral acquisition after HIV exposure. We have recently developed an immunogen (CaV11), tandemly connected overlapping 11-mer peptides spanning the simian immunodeficiency virus (SIV) Gag capsid and Vif proteins, to selectively induce Gag- and Vif-specific CD8+ T cells but not CD4+ T cells. Here, we show protective efficacy of a CaV11-expressing vaccine against repeated intrarectal low-dose SIVmac239 challenge in rhesus macaques. Eight of the twelve vaccinated macaques were protected after eight challenges. Kaplan-Meier analysis indicated significant protection in the vaccinees compared to the unvaccinated macaques. Vaccine-induced Gag-specific CD8+ T cell responses were significantly higher in the protected than the unprotected vaccinees. These results suggest that classical CD8+ T cell induction by viral Env-independent vaccination can confer protection from intrarectal SIV acquisition, highlighting the rationale for this immunogen design to induce virus-specific CD8+ T cells but not CD4+ T cells in HIV-vaccine development.
Collapse
|
4
|
Mouhoub E, Domenech P, Ndao M, Reed MB. The Diverse Applications of Recombinant BCG-Based Vaccines to Target Infectious Diseases Other Than Tuberculosis: An Overview. Front Microbiol 2021; 12:757858. [PMID: 34745066 PMCID: PMC8566895 DOI: 10.3389/fmicb.2021.757858] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Live attenuated Bacillus Calmette-Guérin (BCG) is the world's most widely used vaccine which is mainly administered for its protection against tuberculosis (TB), particularly in young children. However, since its initial use over 100years ago, it has also proven to offer a level of protection against various other pathogens, as a consequence of its non-specific immune enhancing effects. Thus, over the past few decades, recombinant BCG (rBCG) technology has been used as a vector to create rBCG vaccines expressing heterologous antigens that elicit immunity against a range of bacterial, viral, and parasitic diseases. Our goal with this mini-review is to provide an up-to-date survey of the various techniques, approaches, and applications of rBCG-based vaccines for targeting infectious diseases other than TB.
Collapse
Affiliation(s)
- Esma Mouhoub
- The Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
| | - Pilar Domenech
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
| | - Momar Ndao
- The Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
- The Department of Medicine, McGill University, Montreal, QC, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Michael B. Reed
- The Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
- The Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
5
|
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
|
6
|
Baker C, Shi PY. Construction of Stable Reporter Flaviviruses and Their Applications. Viruses 2020; 12:v12101082. [PMID: 32992987 PMCID: PMC7599567 DOI: 10.3390/v12101082] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/28/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Flaviviruses are significant human pathogens that cause frequent emerging and reemerging epidemics around the world. Better molecular tools for studying, diagnosing, and treating these diseases are needed. Reporter viruses represent potent tools to fill this gap but have been hindered by genetic instability. Recent advances have overcome these hurdles, opening the way for increased use of stable reporter flaviviruses to diagnose infections, screen and study antiviral compounds, and serve as potential vaccine vectors.
Collapse
Affiliation(s)
- Coleman Baker
- Microbiology and Immunology Department, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Pei-Yong Shi
- Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Translational Science, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Correspondence:
| |
Collapse
|
7
|
Rosen BC, Pedreño-Lopez N, Ricciardi MJ, Reed JS, Sacha JB, Rakasz EG, Watkins DI. Rhesus Cytomegalovirus-Specific CD8 + Cytotoxic T Lymphocytes Do Not Become Functionally Exhausted in Chronic SIVmac239 Infection. Front Immunol 2020; 11:1960. [PMID: 32922404 PMCID: PMC7457070 DOI: 10.3389/fimmu.2020.01960] [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: 06/08/2020] [Accepted: 07/21/2020] [Indexed: 11/13/2022] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) exert potent antiviral activity after HIV/SIV infection. However, efforts to harness the antiviral efficacy of CTLs for HIV/SIV prophylaxis and therapy have been severely hindered by two major problems: viral escape and exhaustion. By contrast, CTLs directed against human cytomegalovirus (HCMV), a ubiquitous chronic herpesvirus, seldom select for escape mutations and remain functional and refractory to exhaustion during chronic HCMV and HIV infection. Recently, attempts have been made to retarget HCMV-specific CTLs for cancer immunotherapy. We speculate that such a strategy may also be beneficial in the context of HIV/SIV infection, facilitating CTL-mediated control of HIV/SIV replication. As a preliminary assessment of the validity of this approach, we investigated the phenotypes and functionality of rhesus CMV (RhCMV)-specific CTLs in SIVmac239-infected Indian rhesus macaques (RMs), a crucial HIV animal model system. We recently identified two immunodominant, Mamu-A∗02-restricted CTL epitopes derived from RhCMV proteins and sought to evaluate the phenotypic and functional characteristics of these CTL populations in chronic SIVmac239 infection. We analyzed and directly compared RhCMV- and SIVmac239-specific CTLs during SIVmac239 infection in a cohort of Mamu-A∗01 + and Mamu-A∗02 + RMs. CTL populations specific for at least one of the RhCMV-derived CTL epitopes were detected in ten of eleven Mamu-A∗02 + animals tested, and both populations were detected in five of these animals. Neither RhCMV-specific CTL population exhibited significant changes in frequency, memory phenotype, granzyme B expression, exhaustion marker (PD-1 and CTLA-4) expression, or polyfunctionality between pre- and chronic SIVmac239 infection timepoints. In chronic SIVmac239 infection, RhCMV-specific CTLs exhibited higher levels of granzyme B expression and polyfunctionality, and lower levels of exhaustion marker expression, than SIVmac239-specific CTLs. Additionally, compared to SIVmac239-specific CTLs, greater proportions of RhCMV-specific CTLs were of the terminally differentiated effector memory phenotype (CD28- CCR7-) during chronic SIVmac239 infection. These results suggest that, in contrast to SIVmac239-specific CTLs, RhCMV-specific CTLs maintain their phenotypes and cytolytic effector functions during chronic SIVmac239 infection, and that retargeting RhCMV-specific CTLs might be a promising SIV immunotherapeutic strategy.
Collapse
Affiliation(s)
- Brandon C Rosen
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nuria Pedreño-Lopez
- Department of Pathology, George Washington University School of Medicine, Washington, DC, United States
| | - Michael J Ricciardi
- Department of Pathology, George Washington University School of Medicine, Washington, DC, United States
| | - Jason S Reed
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States.,Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States.,Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - David I Watkins
- Department of Pathology, George Washington University School of Medicine, Washington, DC, United States
| |
Collapse
|
8
|
A Novel Immunogen Selectively Eliciting CD8 + T Cells but Not CD4 + T Cells Targeting Immunodeficiency Virus Antigens. J Virol 2020; 94:JVI.01876-19. [PMID: 32024773 DOI: 10.1128/jvi.01876-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/28/2020] [Indexed: 11/20/2022] Open
Abstract
Optimization of immunogen is crucial for induction of effective T-cell responses in the development of a human immunodeficiency virus (HIV) vaccine. Conventional T-cell-based vaccines have been designed to induce virus-specific CD4+ T as well as CD8+ T cells. However, it has been indicated that induction of HIV-specific CD4+ T cells, preferential targets for HIV infection, by vaccination may be detrimental and accelerate viral replication after HIV exposure. In the present study, we present a novel immunogen to selectively induce CD8+ T cells but not CD4+ T cells targeting viral antigens. The immunogen, CaV11, was constructed by tandem connection of overlapping 11-mer peptides spanning simian immunodeficiency virus (SIV) Gag capsid (CA) and Vif. Prime-boost immunization with DNA and Sendai virus (SeV) vectors expressing CaV11 efficiently induced Gag/Vif-specific CD8+ T-cell responses with inefficient Gag/Vif-specific CD4+ T-cell induction in rhesus macaques (n = 6). None of the macaques exhibited the enhancement of acute viral replication after an intravenous high-dose SIV challenge, which was observed in those immunized with DNA and SeV expressing the whole Gag protein in our previous study. Set point viral control postinfection was associated with SeV-specific CD4+ T-cell responses postimmunization, suggesting contribution of SeV-specific helper responses to effective Gag/Vif-specific CD8+ T-cell induction by vaccination. This immunogen design could be a promising method for selective induction of effective anti-HIV CD8+ T-cell responses.IMPORTANCE Induction of effective CD8+ T-cell responses is an important HIV vaccine strategy. Several promising vaccine delivery tools have been developed, and immunogen optimization is now crucial for effective T-cell induction. Conventional immunogens have been designed to induce virus-specific CD4+ T cells as well as CD8+ T cells, but induction of virus-specific CD4+ T cells that are preferential targets for HIV infection could enhance acute HIV proliferation. Here, we designed a novel immunogen to induce HIV-specific CD8+ T cells without HIV-specific CD4+ T-cell induction but with non-HIV antigen-specific CD4+ T-cell help. Our analysis in a macaque AIDS model showed that our immunogen can efficiently elicit effective CD8+ T but not CD4+ T cells targeting viral antigens, resulting in no enhancement of acute viral replication after virus exposure. This immunogen design, also applicable for other currently developed immunogens, could be a promising method for selective induction of effective anti-HIV CD8+ T-cell responses.
Collapse
|
9
|
Martins MA, Gonzalez-Nieto L, Shin YC, Domingues A, Gutman MJ, Maxwell HS, Magnani DM, Ricciardi MJ, Pedreño-Lopez N, Bailey VK, Altman JD, Parks CL, Allison DB, Ejima K, Rakasz EG, Capuano S, Desrosiers RC, Lifson JD, Watkins DI. The Frequency of Vaccine-Induced T-Cell Responses Does Not Predict the Rate of Acquisition after Repeated Intrarectal SIVmac239 Challenges in Mamu-B*08+ Rhesus Macaques. J Virol 2019; 93:e01626-18. [PMID: 30541854 PMCID: PMC6384082 DOI: 10.1128/jvi.01626-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/04/2018] [Indexed: 01/01/2023] Open
Abstract
Approximately 50% of rhesus macaques (RMs) expressing the major histocompatibility complex class I (MHC-I) allele Mamu-B*08 spontaneously control chronic-phase viremia after infection with the pathogenic simian immunodeficiency virus mac239 (SIVmac239) clone. CD8+ T-cell responses in these animals are focused on immunodominant Mamu-B*08-restricted SIV epitopes in Vif and Nef, and prophylactic vaccination with these epitopes increases the incidence of elite control in SIVmac239-infected Mamu-B*08-positive (Mamu-B*08+ ) RMs. Here we evaluated if robust vaccine-elicited CD8+ T-cell responses against Vif and Nef can prevent systemic infection in Mamu-B*08+ RMs following mucosal SIV challenges. Ten Mamu-B*08+ RMs were vaccinated with a heterologous prime/boost/boost regimen encoding Vif and Nef, while six sham-vaccinated MHC-I-matched RMs served as the controls for this experiment. Vaccine-induced CD8+ T cells against Mamu-B*08-restricted SIV epitopes reached high frequencies in blood but were present at lower levels in lymph node and gut biopsy specimens. Following repeated intrarectal challenges with SIVmac239, all control RMs became infected by the sixth SIV exposure. By comparison, four vaccinees were still uninfected after six challenges, and three of them remained aviremic after 3 or 4 additional challenges. The rate of SIV acquisition in the vaccinees was numerically lower (albeit not statistically significantly) than that in the controls. However, peak viremia was significantly reduced in infected vaccinees compared to control animals. We found no T-cell markers that distinguished vaccinees that acquired SIV infection from those that did not. Additional studies will be needed to validate these findings and determine if cellular immunity can be harnessed to prevent the establishment of productive immunodeficiency virus infection.IMPORTANCE It is generally accepted that the antiviral effects of vaccine-induced classical CD8+ T-cell responses against human immunodeficiency virus (HIV) are limited to partial reductions in viremia after the establishment of productive infection. Here we show that rhesus macaques (RMs) vaccinated with Vif and Nef acquired simian immunodeficiency virus (SIV) infection at a lower (albeit not statistically significant) rate than control RMs following repeated intrarectal challenges with a pathogenic SIV clone. All animals in the present experiment expressed the elite control-associated major histocompatibility complex class I (MHC-I) molecule Mamu-B*08 that binds immunodominant epitopes in Vif and Nef. Though preliminary, these results provide tantalizing evidence that the protective efficacy of vaccine-elicited CD8+ T cells may be greater than previously thought. Future studies should examine if vaccine-induced cellular immunity can prevent systemic viral replication in RMs that do not express MHC-I alleles associated with elite control of SIV infection.
Collapse
Affiliation(s)
| | | | - Young C Shin
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Aline Domingues
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Martin J Gutman
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Helen S Maxwell
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Diogo M Magnani
- Department of Pathology, University of Miami, Miami, Florida, USA
| | | | | | - Varian K Bailey
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - John D Altman
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
| | - Christopher L Parks
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, USA
| | - David B Allison
- School of Public Health, Indiana University-Bloomington, Bloomington, Indiana, USA
| | - Keisuke Ejima
- School of Public Health, Indiana University-Bloomington, Bloomington, Indiana, USA
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - David I Watkins
- Department of Pathology, University of Miami, Miami, Florida, USA
| |
Collapse
|
10
|
Kilpeläinen A, Maya-Hoyos M, Saubí N, Soto CY, Joseph Munne J. Advances and challenges in recombinant Mycobacterium bovis BCG-based HIV vaccine development: lessons learned. Expert Rev Vaccines 2018; 17:1005-1020. [PMID: 30300040 DOI: 10.1080/14760584.2018.1534588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Human Immunodeficiency Virus/Acquired Immune Deficiency Syndrome, tuberculosis, and malaria are responsible for most human deaths produced by infectious diseases worldwide. Vaccination against HIV requires generation of memory T cells and neutralizing antibodies, mucosal immunity, and stimulation of an innate immune responses. In this context, the use of Mycobacterium bovis bacillus Calmette-Guérin (BCG) as a live vaccine vehicle is a promising approach for T-cell induction. AREAS COVERED In this review, we provide a comprehensive summary of the literature regarding immunogenicity studies in animal models performed since 2005. Furthermore, we provide expert commentary and 5-year view on how the development of potential recombinant BCG-based HIV vaccines involves careful selection of the HIV antigen, expression vectors, promoters, BCG strain, preclinical animal models, influence of preexisting immunity, and safety issues, for the rational design of recombinant BCG:HIV vaccines to prevent HIV transmission in the general population. EXPERT COMMENTARY The three critical issues to be considered when developing a rBCG:HIV vaccine are codon optimization, antigen localization, and plasmid stability in vivo. The use of integrative expression vectors are likely to improve the mycobacterial vaccine stability 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)
- Athina Kilpeläinen
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Milena Maya-Hoyos
- b Chemistry Department, Faculty of Sciences , Universidad Nacional de Colombia, Ciudad Universitaria , Bogotá , Colombia
| | - Narcís Saubí
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Carlos Y Soto
- b Chemistry Department, Faculty of Sciences , Universidad Nacional de Colombia, Ciudad Universitaria , Bogotá , Colombia
| | - Joan Joseph Munne
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| |
Collapse
|
11
|
Martins MA, Tully DC, Pedreño-Lopez N, von Bredow B, Pauthner MG, Shin YC, Yuan M, Lima NS, Bean DJ, Gonzalez-Nieto L, Domingues A, Gutman MJ, Maxwell HS, Magnani DM, Ricciardi MJ, Bailey VK, Altman JD, Burton DR, Ejima K, Allison DB, Evans DT, Rakasz EG, Parks CL, Bonaldo MC, Capuano S, Lifson JD, Desrosiers RC, Allen TM, Watkins DI. Mamu-B*17+ Rhesus Macaques Vaccinated with env, vif, and nef Manifest Early Control of SIVmac239 Replication. J Virol 2018; 92:e00690-18. [PMID: 29875239 PMCID: PMC6069176 DOI: 10.1128/jvi.00690-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/28/2018] [Indexed: 12/22/2022] Open
Abstract
Certain major histocompatibility complex class I (MHC-I) alleles are associated with spontaneous control of viral replication in human immunodeficiency virus (HIV)-infected people and simian immunodeficiency virus (SIV)-infected rhesus macaques (RMs). These cases of "elite" control of HIV/SIV replication are often immune-mediated, thereby providing a framework for studying anti-lentiviral immunity. In this study, we examined how vaccination impacts SIV replication in RMs expressing the MHC-I allele Mamu-B*17 Approximately 21% of Mamu-B*17+ and 50% of Mamu-B*08+ RMs control chronic-phase viremia after SIVmac239 infection. Because CD8+ T cells targeting Mamu-B*08-restricted SIV epitopes have been implicated in virologic suppression in Mamu-B*08+ RMs, we investigated whether this might also be true for Mamu-B*17+ RMs. Two groups of Mamu-B*17+ RMs were vaccinated with genes encoding Mamu-B*17-restricted epitopes in Vif and Nef. These genes were delivered by themselves (group 1) or together with env (group 2). Group 3 included MHC-I-matched RMs and served as the control group. Surprisingly, the group 1 vaccine regimen had little effect on viral replication compared to group 3, suggesting that unlike Mamu-B*08+ RMs, preexisting SIV-specific CD8+ T cells alone do not facilitate long-term virologic suppression in Mamu-B*17+ RMs. Remarkably, however, 5/8 group 2 vaccinees controlled viremia to <15 viral RNA copies/ml soon after infection. No serological neutralizing activity against SIVmac239 was detected in group 2, although vaccine-elicited gp140-binding antibodies correlated inversely with nadir viral loads. Collectively, these data shed new light on the unique mechanism of elite control in Mamu-B*17+ RMs and implicate vaccine-induced, nonneutralizing anti-Env antibodies in the containment of immunodeficiency virus infection.IMPORTANCE A better understanding of the immune correlates of protection against HIV might facilitate the development of a prophylactic vaccine. Therefore, we investigated simian immunodeficiency virus (SIV) infection outcomes in rhesus macaques expressing the major histocompatibility complex class I allele Mamu-B*17 Approximately 21% of Mamu-B*17+ macaques spontaneously controlled chronic phase viremia after SIV infection, an effect that may involve CD8+ T cells targeting Mamu-B*17-restricted SIV epitopes. We vaccinated Mamu-B*17+ macaques with genes encoding immunodominant epitopes in Vif and Nef alone (group 1) or together with env (group 2). Although neither vaccine regimen prevented SIV infection, 5/8 group 2 vaccinees controlled viremia to below detection limits shortly after infection. This outcome, which was not observed in group 1, was associated with vaccine-induced, nonneutralizing Env-binding antibodies. Together, these findings suggest a limited contribution of Vif- and Nef-specific CD8+ T cells for virologic control in Mamu-B*17+ macaques and implicate anti-Env antibodies in containment of SIV infection.
Collapse
Affiliation(s)
| | - Damien C Tully
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Benjamin von Bredow
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Matthias G Pauthner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, USA
| | - Young C Shin
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Maoli Yuan
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, USA
| | - Noemia S Lima
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz-FIOCRUZ, Rio de Janeiro, Brazil
| | - David J Bean
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Aline Domingues
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Martin J Gutman
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Helen S Maxwell
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - Diogo M Magnani
- Department of Pathology, University of Miami, Miami, Florida, USA
| | | | - Varian K Bailey
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - John D Altman
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
| | - Dennis R Burton
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, California, USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, USA
| | - Keisuke Ejima
- School of Public Health, Indiana University Bloomington, Bloomington, Indiana, USA
| | - David B Allison
- School of Public Health, Indiana University Bloomington, Bloomington, Indiana, USA
| | - David T Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Christopher L Parks
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, USA
| | - Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz-FIOCRUZ, Rio de Janeiro, Brazil
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | | | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - David I Watkins
- Department of Pathology, University of Miami, Miami, Florida, USA
| |
Collapse
|
12
|
Martins MA, Watkins DI. What Is the Predictive Value of Animal Models for Vaccine Efficacy in Humans? Rigorous Simian Immunodeficiency Virus Vaccine Trials Can Be Instructive. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029504. [PMID: 28348034 DOI: 10.1101/cshperspect.a029504] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Simian immunodeficiency virus (SIV) challenge of rhesus macaques provides an invaluable tool to evaluate the clinical prospects of HIV-1 vaccine concepts. However, as with any animal model of human disease, it is crucial to understand the advantages and limitations of this system to maximize the translational value of SIV vaccine studies. Here, we discuss the importance of assessing the efficacy of vaccine prototypes using stringent SIV challenge regimens that mimic HIV-1 transmission and pathogenesis. We also review some of the cautionary tales of HIV-1 vaccine research because they provide general lessons for the preclinical assessment of vaccine candidates.
Collapse
Affiliation(s)
| | - David I Watkins
- Department of Pathology, University of Miami, Miami, Florida 33136
| |
Collapse
|
13
|
Yu JS, Liao HX, Pritchett J, Bowman C, Vivian C, Parks R, Xia SM, Cooper M, Williams WB, Bonsignori M, Reed SG, Chen M, Vandergrift N, Rice CM, Haynes BF. Development of a recombinant yellow fever vector expressing a HIV clade C founder envelope gp120. J Virol Methods 2017; 249:85-93. [PMID: 28837840 DOI: 10.1016/j.jviromet.2017.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 11/29/2022]
Abstract
Development of a HIV-1 vaccine is a major global priority. The yellow fever virus (YFV) attenuated vaccine 17D is among the most effective of currently used vaccines. However, the stability of the YFV17D vector when carrying non-flavivirus genes has been problematic. We have constructed and expressed HIV-1 Env in YFV17D with either single transmembrane (STM) or double transmembrane (DTM) YFV E protein domains for the development of anti-HIV antibodies. Here we describe modifications of the YFV17D vector such that HIV-1 Env gp120 is expressed in up to 5 passages in Vero cells. Immunization with recombinant YFV17D vector prime followed by HIV-1 CH505 TF gp120 protein boosts were able to induce neutralizing antibodies for a HIV-1 tier 1 isolate in mice. This modified YFV vector may be a starting point for constructing HIV-1 vaccine candidate priming vectors.
Collapse
Affiliation(s)
- Jae-Sung Yu
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States.
| | - Hua-Xin Liao
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Jamie Pritchett
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Cindy Bowman
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Callie Vivian
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Robert Parks
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Shi-Mao Xia
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Melissa Cooper
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Wilton B Williams
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Mattia Bonsignori
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA 98102, United States
| | - Meng Chen
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Nathan Vandergrift
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, United States
| | - Barton F Haynes
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
14
|
Martins MA, Tully DC, Shin YC, Gonzalez-Nieto L, Weisgrau KL, Bean DJ, Gadgil R, Gutman MJ, Domingues A, Maxwell HS, Magnani DM, Ricciardi M, Pedreño-Lopez N, Bailey V, Cruz MA, Lima NS, Bonaldo MC, Altman JD, Rakasz E, Capuano S, Reimann KA, Piatak M, Lifson JD, Desrosiers RC, Allen TM, Watkins DI. Rare Control of SIVmac239 Infection in a Vaccinated Rhesus Macaque. AIDS Res Hum Retroviruses 2017; 33:843-858. [PMID: 28503929 DOI: 10.1089/aid.2017.0046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Effector memory T cell (TEM) responses display potent antiviral properties and have been linked to stringent control of simian immunodeficiency virus (SIV) replication. Since recurrent antigen stimulation drives the differentiation of CD8+ T cells toward the TEM phenotype, in this study we incorporated a persistent herpesviral vector into a heterologous prime/boost/boost vaccine approach to maximize the induction of TEM responses. This new regimen resulted in CD8+ TEM-biased responses in four rhesus macaques, three of which controlled viral replication to <1,000 viral RNA copies/ml of plasma for more than 6 months after infection with SIVmac239. Over the course of this study, we made a series of interesting observations in one of these successful controller animals. Indeed, in vivo elimination of CD8αβ+ T cells using a new CD8β-depleting antibody did not abrogate virologic control in this monkey. Only after its CD8α+ lymphocytes were depleted did SIV rebound, suggesting that CD8αα+ but not CD8αβ+ cells were controlling viral replication. By 2 weeks postinfection (PI), the only SIV sequences that could be detected in this animal harbored a small in-frame deletion in nef affecting six amino acids. Deep sequencing of the SIVmac239 challenge stock revealed no evidence of this polymorphism. However, sequencing of the rebound virus following CD8α depletion at week 38.4 PI again revealed only the six-amino acid deletion in nef. While any role for immunological pressure on the selection of this deleted variant remains uncertain, our data provide anecdotal evidence that control of SIV replication can be maintained without an intact CD8αβ+ T cell compartment.
Collapse
Affiliation(s)
| | - Damien C. Tully
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | - Young C. Shin
- Department of Pathology, University of Miami, Miami, Florida
| | | | - Kim L. Weisgrau
- Wisconsin National Primate Research Center, University of Wisconsin—Madison, Madison, Wisconsin
| | - David J. Bean
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | - Rujuta Gadgil
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | | | - Aline Domingues
- Department of Pathology, University of Miami, Miami, Florida
| | | | | | | | | | - Varian Bailey
- Department of Pathology, University of Miami, Miami, Florida
| | - Michael A. Cruz
- Department of Pathology, University of Miami, Miami, Florida
| | - Noemia S. Lima
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz–FIOCRUZ, Rio de Janeiro, Brazil
| | - Myrna C. Bonaldo
- Laboratório de Biologia Molecular de Flavivirus, Instituto Oswaldo Cruz–FIOCRUZ, Rio de Janeiro, Brazil
| | - John D. Altman
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia
| | - Eva Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin—Madison, Madison, Wisconsin
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin—Madison, Madison, Wisconsin
| | - Keith A. Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | - Todd M. Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | | |
Collapse
|
15
|
Martins MA, Shin YC, Gonzalez-Nieto L, Domingues A, Gutman MJ, Maxwell HS, Castro I, Magnani DM, Ricciardi M, Pedreño-Lopez N, Bailey V, Betancourt D, Altman JD, Pauthner M, Burton DR, von Bredow B, Evans DT, Yuan M, Parks CL, Ejima K, Allison DB, Rakasz E, Barber GN, Capuano S, Lifson JD, Desrosiers RC, Watkins DI. Vaccine-induced immune responses against both Gag and Env improve control of simian immunodeficiency virus replication in rectally challenged rhesus macaques. PLoS Pathog 2017; 13:e1006529. [PMID: 28732035 PMCID: PMC5540612 DOI: 10.1371/journal.ppat.1006529] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/02/2017] [Accepted: 07/13/2017] [Indexed: 01/28/2023] Open
Abstract
The ability to control lentivirus replication may be determined, in part, by the extent to which individual viral proteins are targeted by the immune system. Consequently, defining the antigens that elicit the most protective immune responses may facilitate the design of effective HIV-1 vaccines. Here we vaccinated four groups of rhesus macaques with a heterologous vector prime/boost/boost/boost (PBBB) regimen expressing the following simian immunodeficiency virus (SIV) genes: env, gag, vif, rev, tat, and nef (Group 1); env, vif, rev, tat, and nef (Group 2); gag, vif, rev, tat, and nef (Group 3); or vif, rev, tat, and nef (Group 4). Following repeated intrarectal challenges with a marginal dose of the neutralization-resistant SIVmac239 clone, vaccinees in Groups 1-3 became infected at similar rates compared to control animals. Unexpectedly, vaccinees in Group 4 became infected at a slower pace than the other animals, although this difference was not statistically significant. Group 1 exhibited the best post-acquisition virologic control of SIV infection, with significant reductions in both peak and chronic phase viremia. Indeed, 5/8 Group 1 vaccinees had viral loads of less than 2,000 vRNA copies/mL of plasma in the chronic phase. Vaccine regimens that did not contain gag (Group 2), env (Group 3), or both of these inserts (Group 4) were largely ineffective at decreasing viremia. Thus, vaccine-induced immune responses against both Gag and Env appeared to maximize control of immunodeficiency virus replication. Collectively, these findings are relevant for HIV-1 vaccine design as they provide additional insights into which of the lentiviral proteins might serve as the best vaccine immunogens.
Collapse
Affiliation(s)
- Mauricio A. Martins
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Young C. Shin
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Lucas Gonzalez-Nieto
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Aline Domingues
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Martin J. Gutman
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Helen S. Maxwell
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Iris Castro
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Diogo M. Magnani
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Michael Ricciardi
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Nuria Pedreño-Lopez
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Varian Bailey
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - Dillon Betancourt
- Department of Microbiology and Immunology, University of Miami, Miami, Florida, United States of America
| | - John D. Altman
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, United States of America
| | - Matthias Pauthner
- Department of Immunology and Microbiology, IAVI Neutralizing Antibody Center, Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, United States of America
| | - Dennis R. Burton
- Department of Immunology and Microbiology, IAVI Neutralizing Antibody Center, Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), The Scripps Research Institute, La Jolla, California, United States of America
| | - Benjamin von Bredow
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - David T. Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Maoli Yuan
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Christopher L. Parks
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Keisuke Ejima
- Section on Statistical Genetics, Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David B. Allison
- Section on Statistical Genetics, Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Eva Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Glen N. Barber
- Department of Cell Biology, University of Miami, Miami, Florida, United States of America
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Ronald C. Desrosiers
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| | - David I. Watkins
- Department of Pathology, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
16
|
Spanevello F, Calistri A, Del Vecchio C, Mantelli B, Frasson C, Basso G, Palù G, Cavazzana M, Parolin C. Development of Lentiviral Vectors Simultaneously Expressing Multiple siRNAs Against CCR5, vif and tat/rev Genes for an HIV-1 Gene Therapy Approach. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e312. [PMID: 27093170 PMCID: PMC5014525 DOI: 10.1038/mtna.2016.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/08/2016] [Indexed: 02/08/2023]
Abstract
Gene therapy holds considerable promise for the functional cure of HIV-1 infection and, in this context, RNA interference (RNAi)-based approaches represent powerful strategies. Stable expression of small interfering RNAs (siRNAs) targeting HIV genes or cellular cofactors has the potential to render HIV-1 susceptible cells resistant to infection. To inhibit different steps of virus life cycle, self-inactivating lentiviral vectors expressing multiple siRNAs targeting the CCR5 cellular gene as well as vif and tat/rev viral transcripts, under the control of different RNA polymerase III promoters (U6, 7SK, H1) were developed. The use of a single RNA polymerase III promoter driving the expression of a sequence giving rise to three siRNAs directed against the selected targets (e-shRNA) was also investigated. Luciferase assay and inhibition of HIV-1 replication in human Jurkat T-cell line were adopted to select the best combination of promoter/siRNA. The efficacy of selected developed combinatorial vectors in interfering with viral replication was evaluated in human primary CD4(+) T lymphocytes. We identified two effective anti-HIV combinatorial vectors that conferred protection against R5- and X4- tropic viruses. Overall, our results showed that the antiviral effect is influenced by different factors, including the promoter used to express the RNAi molecules and the selected cassette combination. These findings contribute to gain further insights in the design of RNAi-based gene therapy approaches against HIV-1 for clinical application.
Collapse
Affiliation(s)
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Barbara Mantelli
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Chiara Frasson
- Oncohematology Laboratory, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Città della Speranza (IRP), Padova, Italy
| | - Giuseppe Basso
- Oncohematology Laboratory, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marina Cavazzana
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique–Hôpitaux de Paris, INSERM, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
- INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
17
|
Fuchs SP, Martinez-Navio JM, Piatak M, Lifson JD, Gao G, Desrosiers RC. AAV-Delivered Antibody Mediates Significant Protective Effects against SIVmac239 Challenge in the Absence of Neutralizing Activity. PLoS Pathog 2015; 11:e1005090. [PMID: 26248318 PMCID: PMC4527674 DOI: 10.1371/journal.ppat.1005090] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/20/2015] [Indexed: 01/12/2023] Open
Abstract
Long-term delivery of potent broadly-neutralizing antibodies is a promising approach for the prevention of HIV-1 infection. We used AAV vector intramuscularly to deliver anti-SIV monoclonal antibodies (mAbs) in IgG1 form to rhesus monkeys. Persisting levels of delivered mAb as high as 270 μg/ml were achieved. However, host antibody responses to the delivered antibody were observed in 9 of the 12 monkeys and these appeared to limit the concentration of delivered antibody that could be achieved. This is reflected in the wide range of delivered mAb concentrations that were achieved: 1-270 μg/ml. Following repeated, marginal dose, intravenous challenge with the difficult-to-neutralize SIVmac239, the six monkeys in the AAV-5L7 IgG1 mAb group showed clear protective effects despite the absence of detectable neutralizing activity against the challenge virus. The protective effects included: lowering of viral load at peak height; lowering of viral load at set point; delay in the time to peak viral load from the time of the infectious virus exposure. All of these effects were statistically significant. In addition, the monkey with the highest level of delivered 5L7 mAb completely resisted six successive SIVmac239 i.v. challenges, including a final challenge with a dose of 10 i.v. infectious units. Our results demonstrate the continued promise of this approach for the prevention of HIV-1 infection in people. However, the problem of anti-antibody responses will need to be understood and overcome for the promise of this approach to be effectively realized.
Collapse
Affiliation(s)
- Sebastian P. Fuchs
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - José M. Martinez-Navio
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ronald C. Desrosiers
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
18
|
Mothe B, Hu X, Llano A, Rosati M, Olvera A, Kulkarni V, Valentin A, Alicea C, Pilkington GR, Sardesai NY, Rocafort M, Crespo M, Carrillo J, Marco A, Mullins JI, Dorrell L, Hanke T, Clotet B, Pavlakis GN, Felber BK, Brander C. A human immune data-informed vaccine concept elicits strong and broad T-cell specificities associated with HIV-1 control in mice and macaques. J Transl Med 2015; 13:60. [PMID: 25879820 PMCID: PMC4336696 DOI: 10.1186/s12967-015-0392-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 01/13/2015] [Indexed: 12/03/2022] Open
Abstract
Background None of the HIV T-cell vaccine candidates that have reached advanced clinical testing have been able to induce protective T cell immunity. A major reason for these failures may have been suboptimal T cell immunogen designs. Methods To overcome this problem, we used a novel immunogen design approach that is based on functional T cell response data from more than 1,000 HIV-1 clade B and C infected individuals and which aims to direct the T cell response to the most vulnerable sites of HIV-1. Results Our approach identified 16 regions in Gag, Pol, Vif and Nef that were relatively conserved and predominantly targeted by individuals with reduced viral loads. These regions formed the basis of the HIVACAT T-cell Immunogen (HTI) sequence which is 529 amino acids in length, includes more than 50 optimally defined CD4+ and CD8+ T-cell epitopes restricted by a wide range of HLA class I and II molecules and covers viral sites where mutations led to a dramatic reduction in viral replicative fitness. In both, C57BL/6 mice and Indian rhesus macaques immunized with an HTI-expressing DNA plasmid (DNA.HTI) induced broad and balanced T-cell responses to several segments within Gag, Pol, and Vif. DNA.HTI induced robust CD4+ and CD8+ T cell responses that were increased by a booster vaccination using modified virus Ankara (MVA.HTI), expanding the DNA.HTI induced response to up to 3.2% IFN-γ T-cells in macaques. HTI-specific T cells showed a central and effector memory phenotype with a significant fraction of the IFN-γ+ CD8+ T cells being Granzyme B+ and able to degranulate (CD107a+). Conclusions These data demonstrate the immunogenicity of a novel HIV-1 T cell vaccine concept that induced broadly balanced responses to vulnerable sites of HIV-1 while avoiding the induction of responses to potential decoy targets that may divert effective T-cell responses towards variable and less protective viral determinants. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0392-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Beatriz Mothe
- IrsiCaixa AIDS Research Institute - HIVACAT, Hospital Germans Trias i Pujol, Crta Canyet s/n., 08916, Badalona, Barcelona, Spain. .,'Lluita contra la Sida' Foundation, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain. .,Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain.
| | - Xintao Hu
- Human Retrovirus Pathogenesis Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Anuska Llano
- IrsiCaixa AIDS Research Institute - HIVACAT, Hospital Germans Trias i Pujol, Crta Canyet s/n., 08916, Badalona, Barcelona, Spain.
| | - Margherita Rosati
- Human Retrovirus Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Alex Olvera
- IrsiCaixa AIDS Research Institute - HIVACAT, Hospital Germans Trias i Pujol, Crta Canyet s/n., 08916, Badalona, Barcelona, Spain.
| | - Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Antonio Valentin
- Human Retrovirus Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Guy R Pilkington
- Human Retrovirus Pathogenesis Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | | | - Muntsa Rocafort
- IrsiCaixa AIDS Research Institute - HIVACAT, Hospital Germans Trias i Pujol, Crta Canyet s/n., 08916, Badalona, Barcelona, Spain.
| | - Manel Crespo
- HIV Unit, Hospital de la Vall d'Hebrón, Barcelona, Spain.
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute - HIVACAT, Hospital Germans Trias i Pujol, Crta Canyet s/n., 08916, Badalona, Barcelona, Spain.
| | | | | | - Lucy Dorrell
- Nuffield Department of Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, UK.
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, UK.
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute - HIVACAT, Hospital Germans Trias i Pujol, Crta Canyet s/n., 08916, Badalona, Barcelona, Spain. .,'Lluita contra la Sida' Foundation, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain. .,Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain. .,Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - George N Pavlakis
- Human Retrovirus Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, National Cancer Institute-Frederick, Frederick, MD, USA.
| | - Christian Brander
- IrsiCaixa AIDS Research Institute - HIVACAT, Hospital Germans Trias i Pujol, Crta Canyet s/n., 08916, Badalona, Barcelona, Spain. .,Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain. .,Universitat Autònoma de Barcelona, Barcelona, Spain. .,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
19
|
Valentin A, McKinnon K, Li J, Rosati M, Kulkarni V, Pilkington GR, Bear J, Alicea C, Vargas-Inchaustegui DA, Jean Patterson L, Pegu P, Liyanage NPM, Gordon SN, Vaccari M, Wang Y, Hogg AE, Frey B, Sui Y, Reed SG, Sardesai NY, Berzofsky JA, Franchini G, Robert-Guroff M, Felber BK, Pavlakis GN. Comparative analysis of SIV-specific cellular immune responses induced by different vaccine platforms in rhesus macaques. Clin Immunol 2014; 155:91-107. [PMID: 25229164 DOI: 10.1016/j.clim.2014.09.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 12/21/2022]
Abstract
To identify the most promising vaccine candidates for combinatorial strategies, we compared five SIV vaccine platforms including recombinant canary pox virus ALVAC, replication-competent adenovirus type 5 host range mutant RepAd, DNA, modified vaccinia Ankara (MVA), peptides and protein in distinct combinations. Three regimens used viral vectors (prime or boost) and two regimens used plasmid DNA. Analysis at necropsy showed that the DNA-based vaccine regimens elicited significantly higher cellular responses against Gag and Env than any of the other vaccine platforms. The T cell responses induced by most vaccine regimens disseminated systemically into secondary lymphoid tissues (lymph nodes, spleen) and effector anatomical sites (including liver, vaginal tissue), indicative of their role in viral containment at the portal of entry. The cellular and reported humoral immune response data suggest that combination of DNA and viral vectors elicits a balanced immunity with strong and durable responses able to disseminate into relevant mucosal sites.
Collapse
Affiliation(s)
- Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Katherine McKinnon
- FACS Core Facility, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jinyao Li
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Guy R Pilkington
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Diego A Vargas-Inchaustegui
- Immune Biology of Retroviral Infection Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - L Jean Patterson
- Immune Biology of Retroviral Infection Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Poonam Pegu
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Namal P M Liyanage
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Shari N Gordon
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Monica Vaccari
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yichuan Wang
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Alison E Hogg
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Blake Frey
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yongjun Sui
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Steven G Reed
- Infectious Diseases Research Institute, Seattle, WA, USA
| | | | - Jay A Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Marjorie Robert-Guroff
- Immune Biology of Retroviral Infection Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|