1
|
Bubar KM, Reinholt K, Kissler SM, Lipsitch M, Cobey S, Grad YH, Larremore DB. Model-informed COVID-19 vaccine prioritization strategies by age and serostatus. Science 2021; 371:916-921. [PMID: 33479118 DOI: 10.1126/science:abe6959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/12/2021] [Indexed: 05/25/2023]
Abstract
Limited initial supply of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine raises the question of how to prioritize available doses. We used a mathematical model to compare five age-stratified prioritization strategies. A highly effective transmission-blocking vaccine prioritized to adults ages 20 to 49 years minimized cumulative incidence, but mortality and years of life lost were minimized in most scenarios when the vaccine was prioritized to adults greater than 60 years old. Use of individual-level serological tests to redirect doses to seronegative individuals improved the marginal impact of each dose while potentially reducing existing inequities in COVID-19 impact. Although maximum impact prioritization strategies were broadly consistent across countries, transmission rates, vaccination rollout speeds, and estimates of naturally acquired immunity, this framework can be used to compare impacts of prioritization strategies across contexts.
Collapse
Affiliation(s)
- Kate M Bubar
- Department of Applied Mathematics, University of Colorado Boulder, Boulder, CO 80309, USA.
- IQ Biology Program, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Kyle Reinholt
- Department of Computer Science, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Stephen M Kissler
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Marc Lipsitch
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- Center for Communicable Disease Dynamics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL 60637, USA
| | - Yonatan H Grad
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Daniel B Larremore
- Department of Computer Science, University of Colorado Boulder, Boulder, CO 80309, USA.
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
2
|
Bubar KM, Reinholt K, Kissler SM, Lipsitch M, Cobey S, Grad YH, Larremore DB. Model-informed COVID-19 vaccine prioritization strategies by age and serostatus. Science 2021; 371:916-921. [PMID: 33479118 PMCID: PMC7963218 DOI: 10.1126/science.abe6959] [Citation(s) in RCA: 444] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
Limited initial supply of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine raises the question of how to prioritize available doses. We used a mathematical model to compare five age-stratified prioritization strategies. A highly effective transmission-blocking vaccine prioritized to adults ages 20 to 49 years minimized cumulative incidence, but mortality and years of life lost were minimized in most scenarios when the vaccine was prioritized to adults greater than 60 years old. Use of individual-level serological tests to redirect doses to seronegative individuals improved the marginal impact of each dose while potentially reducing existing inequities in COVID-19 impact. Although maximum impact prioritization strategies were broadly consistent across countries, transmission rates, vaccination rollout speeds, and estimates of naturally acquired immunity, this framework can be used to compare impacts of prioritization strategies across contexts.
Collapse
Affiliation(s)
- Kate M Bubar
- Department of Applied Mathematics, University of Colorado Boulder, Boulder, CO 80309, USA.
- IQ Biology Program, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Kyle Reinholt
- Department of Computer Science, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Stephen M Kissler
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Marc Lipsitch
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- Center for Communicable Disease Dynamics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL 60637, USA
| | - Yonatan H Grad
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Daniel B Larremore
- Department of Computer Science, University of Colorado Boulder, Boulder, CO 80309, USA.
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
3
|
Bubar KM, Reinholt K, Kissler SM, Lipsitch M, Cobey S, Grad YH, Larremore DB. Model-informed COVID-19 vaccine prioritization strategies by age and serostatus. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2020.09.08.20190629. [PMID: 33330882 PMCID: PMC7743091 DOI: 10.1101/2020.09.08.20190629] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Limited initial supply of SARS-CoV-2 vaccine raises the question of how to prioritize available doses. Here, we used a mathematical model to compare five age-stratified prioritization strategies. A highly effective transmission-blocking vaccine prioritized to adults ages 20-49 years minimized cumulative incidence, but mortality and years of life lost were minimized in most scenarios when the vaccine was prioritized to adults over 60 years old. Use of individual-level serological tests to redirect doses to seronegative individuals improved the marginal impact of each dose while potentially reducing existing inequities in COVID-19 impact. While maximum impact prioritization strategies were broadly consistent across countries, transmission rates, vaccination rollout speeds, and estimates of naturally acquired immunity, this framework can be used to compare impacts of prioritization strategies across contexts.
Collapse
Affiliation(s)
- Kate M. Bubar
- Department of Applied Mathematics, University of Colorado Boulder, Boulder, CO, 80303, USA
- IQ Biology Program, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Kyle Reinholt
- Department of Computer Science, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Stephen M. Kissler
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Marc Lipsitch
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, 60637, USA
| | - Yonatan H. Grad
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Daniel B. Larremore
- Department of Computer Science, University of Colorado Boulder, Boulder, CO, 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
4
|
Chea LS, Amara RR. Immunogenicity and efficacy of DNA/MVA HIV vaccines in rhesus macaque models. Expert Rev Vaccines 2017; 16:973-985. [PMID: 28838267 DOI: 10.1080/14760584.2017.1371594] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Despite 30 years of research on HIV, a vaccine to prevent infection and limit disease progression remains elusive. The RV144 trial showed moderate, but significant protection in humans and highlighted the contribution of antibody responses directed against HIV envelope as an important immune correlate for protection. Efforts to further build upon the progress include the use of a heterologous prime-boost regimen using DNA as the priming agent and the attenuated vaccinia virus, Modified Vaccinia Ankara (MVA), as a boosting vector for generating protective HIV-specific immunity. Areas covered: In this review, we summarize the immunogenicity of DNA/MVA vaccines in non-human primate models and describe the efficacy seen in SIV infection models. We discuss immunological correlates of protection determined by these studies and potential approaches for improving the protective immunity. Additionally, we describe the current progress of DNA/MVA vaccines in human trials. Expert commentary: Efforts over the past decade have provided the opportunity to better understand the dynamics of vaccine-induced immune responses and immune correlates of protection against HIV. Based on what we have learned, we outline multiple areas where the field will likely focus on in the next five years.
Collapse
Affiliation(s)
- Lynette Siv Chea
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| | - Rama Rao Amara
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| |
Collapse
|
5
|
Spieker AJ, Huang Y. A method to address between-subject heterogeneity for identification of principal surrogate markers in repeated low-dose challenge HIV vaccine studies. Stat Med 2017; 36:4071-4080. [PMID: 28758224 DOI: 10.1002/sim.7419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/15/2017] [Accepted: 06/26/2017] [Indexed: 11/08/2022]
Abstract
Repeated low-dose challenge designs in nonhuman primate studies have recently received attention in the literature as a means of evaluating vaccines for HIV prevention and identifying immune surrogates for their protective effects. Existing methods for surrogate identification in this type of study design rely on the assumption of homogeneity across subjects (namely, independent infection risks after each challenge within each subject and conditional on covariates). In practice, random variation across subjects is likely to occur because of unmeasured biologic factors. Failure to account for this heterogeneity or within-subject correlation can result in biased inference regarding the surrogate value of immune biomarkers and underpowered study designs for detecting surrogate endpoints. In this paper, we adopt a discrete-time survival model with random effects to account for between-subject heterogeneity, and we develop estimators and testing procedures for evaluating principal surrogacy of immune biomarkers. Simulation studies reveal that the heterogeneous model achieves substantial bias reduction compared to the homogeneous model, with little cost of efficiency. We recommend the use of this heterogeneous model as a complementary tool to existing methods when designing and analyzing repeated low-dose challenge studies for evaluating surrogate endpoints.
Collapse
Affiliation(s)
- Andrew J Spieker
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
6
|
Parsimonious Determination of the Optimal Infectious Dose of a Pathogen for Nonhuman Primate Models. PLoS Pathog 2015; 11:e1005100. [PMID: 26285041 PMCID: PMC4540461 DOI: 10.1371/journal.ppat.1005100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/20/2015] [Indexed: 11/22/2022] Open
Abstract
The nonhuman primate (NHP) model is often the best experimental model for testing interventions designed to block infection by human pathogens, such as HIV, tuberculosis, and malaria. A physiological model may require the use of a limiting dose of the infectious agent, where only a fraction of animals become infected upon any given challenge. Determining the challenge dose of the pathogen in such experiments is critical to the success of the experiment: using too-high or too-low a challenge dose may lead to false negative results and an excessive use of animals. Here I define an optimized protocol for defining the dose of pathogen that infects 50% of the time (AID50); other challenge doses, e.g. AID80, can be easily calculated from the same data. This protocol minimizes the number of animals, as well as resources and procedures, while providing an estimate of the AID50 within 1.5-fold of the true value. Exposing nonhuman primates to infectious pathogens (such as tuberculosis, malaria, or the simian equivalent of HIV) is an important model for testing vaccines or other interventions designed to prevent infection or disease. In fact, demonstrating efficacy in animals is often a requirement before clinical testing in humans can be started. A critical variable in such testing is the dose of the pathogen used: this dose should be similar to what humans would encounter. Using too-high a dose may overcome the intervention and mask a successful approach; using too-low a dose may not be relevant. Often, an optimal dose will lead to “successful” infections only a fraction of the times the animal is exposed. A successful intervention experiment therefore needs to use a precisely calibrated dose of the infectious agent; this calibration can only be done by exposing animals to a range of doses and measuring how often they become infected. Here I define the most parsimonious method for performing this calibration: one that uses the least number of animals and procedures. Given the large number of new pathogens being tested in animal models, adoption of such a parsimonious protocol is both economically and ethically warranted, and will thereby enable favorable review of proposed animal use numbers by Institutional Animal Care and Use Committees.
Collapse
|
7
|
Nolen TL, Hudgens MG, Senb PK, Koch GG. Analysis of repeated low-dose challenge studies. Stat Med 2015; 34:1981-92. [PMID: 25752266 DOI: 10.1002/sim.6462] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 02/06/2015] [Accepted: 02/10/2015] [Indexed: 01/11/2023]
Abstract
Preclinical evaluation of candidate human immunodeficiency virus (HIV) vaccines entails challenge studies whereby non-human primates such as macaques are vaccinated with either an active or control vaccine and then challenged (exposed) with a simian-version of HIV. Repeated low-dose challenge (RLC) studies in which each macaque is challenged multiple times (either until infection or some maximum number of challenges is reached) are becoming more common in an effort to mimic natural exposure to HIV in humans. Statistical methods typically employed for the testing for a vaccine effect in RLC studies include a modified version of Fisher's exact test as well as large sample approaches such as the usual log-rank test. Unfortunately, these methods are not guaranteed to provide a valid test for the effect of vaccination. On the other hand, valid tests for vaccine effect such as the exact log-rank test may not be easy to implement using software available to many researchers. This paper details which statistical approaches are appropriate for the analysis of RLC studies, and how to implement these methods easily in SAS or R.
Collapse
Affiliation(s)
- Tracy L Nolen
- RTI International, The Research Triangle Park, NC, 27709, U.S.A
| | | | | | | |
Collapse
|
8
|
Long DM, Hudgens MG, Wu CD. Surrogates of protection in repeated low-dose challenge experiments. Stat Med 2015; 34:1747-60. [PMID: 25628249 DOI: 10.1002/sim.6436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 12/22/2014] [Accepted: 01/07/2015] [Indexed: 11/10/2022]
Abstract
A critical step toward developing a successful vaccine to control the human immunodeficiency virus pandemic entails evaluation of vaccine candidates in non-human primates (NHPs). Historically, these studies have usually entailed challenges (i.e., exposures) with very high doses of a simian version of human immunodeficiency virus, resulting in infection of all NHPs in the experiment after a single challenge. More recently, researchers have begun to conduct repeated low-dose challenge (RLC) studies in NHPs that are believed to more closely mimic typical exposure in natural human transmission settings. One objective of RLC studies is to assess whether measured immune responses to vaccination can serve as surrogate endpoints for the primary endpoint of interest, namely infection. In this paper, different designs of RLC studies for assessing a binary surrogate of protection are considered.
Collapse
Affiliation(s)
- Dustin M Long
- Department of Biostatistics, West Virginia University, Morgantown, WV, U.S.A
| | | | | |
Collapse
|
9
|
Regoes RR, Magnus C. The role of chance in primate lentiviral infectivity: from protomer to host organism. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 129:327-51. [PMID: 25595809 DOI: 10.1016/bs.pmbts.2014.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Infection is best described as a stochastic process. Whether a host becomes infected upon exposure has a strong random element. The same applies to cells exposed to virions. In this review, we show how the mathematical formalism for stochastic processes has been used to describe and understand the infection by the Human and Simian Immunodeficiency Virus on different levels. We survey quantitative studies on the establishment of infection in the host (the organismal level) and on the infection of target cells (the cellular and molecular level). We then discuss how a synthesis of the approaches across these levels could give rise to a predictive framework for assessing the efficacy of microbicides and vaccines.
Collapse
Affiliation(s)
- Roland R Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland.
| | - Carsten Magnus
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Kang C, Huang Y, Miller CJ. A discrete-time survival model with random effects for designing and analyzing repeated low-dose challenge experiments. Biostatistics 2014; 16:295-310. [PMID: 25190513 DOI: 10.1093/biostatistics/kxu040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Repeated low-dose (RLD) challenge designs are important in HIV vaccine research. Current methods for RLD designs rely heavily on an assumption of homogeneous risk of infection among animals, which, upon violation, can lead to invalid inferences and underpowered study designs. We propose to fit a discrete-time survival model with random effects that allows for heterogeneity in the risk of infection among animals and allows for predetermined challenge dose changes over time. Based on this model, we derive likelihood ratio tests and estimators for vaccine efficacy. A two-stage approach is proposed for optimizing the RLD design under cost constraints. Simulation studies demonstrate good finite sample properties of the proposed method and its superior performance compared to existing methods. We illustrate the application of the heterogeneous infection risk model on data from a real simian immunodeficiency virus vaccine study using Rhesus Macaques. The results of our study provide useful guidance for future RLD experimental design.
Collapse
Affiliation(s)
- Chaeryon Kang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Christopher J Miller
- Center for Comparative Medicine and California National Primate Research Center, University of California, Davis, CA 95616, USA
| |
Collapse
|
11
|
Yang OO, Ibarrondo FJ, Price C, Hultin LE, Elliott J, Hultin PM, Shih R, Hausner MA, Ng HL, Hoffman J, Jamieson BD, Anton PA. Differential blood and mucosal immune responses against an HIV-1 vaccine administered via inguinal or deltoid injection. PLoS One 2014; 9:e88621. [PMID: 24558403 PMCID: PMC3928250 DOI: 10.1371/journal.pone.0088621] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/01/2014] [Indexed: 12/25/2022] Open
Abstract
Mucosal immunity is central to sexual transmission and overall pathogenesis of HIV-1 infection, but the ability of vaccines to induce immune responses in mucosal tissue compartments is poorly defined. Because macaque vaccine studies suggest that inguinal (versus limb) vaccination may better target sexually-exposed mucosa, we performed a randomized, double-blinded, placebo-controlled Phase I trial in HIV-1-uninfected volunteers, using the recombinant Canarypox (CP) vaccine vCP205 delivered by different routes. 12 persons received vaccine and 6 received placebo, divided evenly between deltoid-intramuscular (deltoid-IM) or inguinal-subcutaneous (inguinal-SC) injection routes. The most significant safety events were injection site reactions (Grade 3) in one inguinal vaccinee. CP-specific antibodies were detected in the blood of all 12 vaccinees by Day 24, while HIV-1-specific antibodies were observed in the blood and gut mucosa of 1/9 and 4/9 evaluated vaccinees respectively, with gut antibodies appearing earlier in inguinal vaccinees (24–180 versus 180–365 days). HIV-1-specific CD8+ T lymphocytes (CTLs) were observed in 7/12 vaccinees, and blood and gut targeting were distinct. Within blood, both deltoid and inguinal responders had detectable CTL responses by 17–24 days; inguinal responders had early responses (within 10 days) while deltoid responders had later responses (24–180 days) in gut mucosa. Our results demonstrate relative safety of inguinal vaccination and qualitative or quantitative compartmentalization of immune responses between blood and gut mucosa, and highlight the importance of not only evaluating early blood responses to HIV-1 vaccines but also mucosal responses over time. Trial Registration ClinicalTrials.gov NCT00076817
Collapse
Affiliation(s)
- Otto O. Yang
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
- AIDS Healthcare Foundation, Los Angeles, California, United States of America
- * E-mail:
| | - F. Javier Ibarrondo
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Charles Price
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Lance E. Hultin
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Julie Elliott
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Patricia M. Hultin
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Roger Shih
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Mary Ann Hausner
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Hwee L. Ng
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Hoffman
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Beth D. Jamieson
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Peter A. Anton
- Department of Medicine and University of California Los Angeles AIDS Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
12
|
Davis D, Koornstra W, Fagrouch Z, Verschoor EJ, Heeney JL, Bogers WMJM. In vitro neutralization of low dose inocula at physiological concentrations of a monoclonal antibody which protects macaques against SHIV challenge. PLoS One 2013; 8:e72702. [PMID: 23977339 PMCID: PMC3745472 DOI: 10.1371/journal.pone.0072702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/10/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Passive transfer of antibodies can be protective in the simian human immunodeficiency virus (SHIV)--rhesus macaque challenge model. The human monoclonal antibody IgG1 b12 neutralizes human immunodeficiency type 1 (HIV-1) in vitro and protects against challenge by SHIV. Our hypothesis is that neutralizing antibodies can only completely inactivate a relatively small number of infectious virus. METHODS AND FINDINGS We have used GHOST cell assays to quantify individual infectious events with HIV-1SF162 and its SHIV derivatives: the relatively neutralization sensitive SHIV(SF162P4) isolate and the more resistant SHIV(SF162P3). A plot of the number of fluorescent GHOST cells with increasing HIV-1SF162 dose is not linear. It is likely that with high-dose inocula, infection with multiple virus produces additive fluorescence in individual cells. In studies of the neutralization kinetics of IgG1 b12 against these isolates, events during the absorption phase of the assay, as well as the incubation phase, determine the level of neutralization. It is possible that complete inactivation of a virus is limited to the time it is exposed on the cell surface. Assays can be modified so that neutralization of these very low doses of virus can be quantified. A higher concentration of antibody is required to neutralize the same dose of resistant SHIV(SF162P3) than the sensitive SHIV(SF162P4). In the absence of selection during passage, the density of the CCR5 co-receptor on the GHOST cell surface is reduced. Changes in the CD4 : CCR5 density ratio influence neutralization. CONCLUSIONS Low concentrations of IgG1 b12 completely inactivate small doses of the neutralization resistant SHIV(SF162P3). Assays need to be modified to quantify this effect. Results from modified assays may predict protection following repeated low-dose shiv challenges in rhesus macaques. It should be possible to induce this level of antibody by vaccination so that modified assays could predict the outcome of human trials.
Collapse
Affiliation(s)
- David Davis
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | | | | | | | |
Collapse
|
13
|
Vasconcelos JR, Dominguez MR, Araújo AF, Ersching J, Tararam CA, Bruna-Romero O, Rodrigues MM. Relevance of long-lived CD8(+) T effector memory cells for protective immunity elicited by heterologous prime-boost vaccination. Front Immunol 2012; 3:358. [PMID: 23264773 PMCID: PMC3525016 DOI: 10.3389/fimmu.2012.00358] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/10/2012] [Indexed: 11/13/2022] Open
Abstract
Owing to the importance of major histocompatibility complex class Ia-restricted CD8(+) T cells for host survival following viral, bacterial, fungal, or parasitic infection, it has become largely accepted that these cells should be considered in the design of a new generation of vaccines. For the past 20 years, solid evidence has been provided that the heterologous prime-boost regimen achieves the best results in terms of induction of long-lived protective CD8(+) T cells against a variety of experimental infections. Although this regimen has often been used experimentally, as is the case for many vaccines, the mechanism behind the efficacy of this vaccination regimen is still largely unknown. The main purpose of this review is to examine the characteristics of the protective CD8(+) T cells generated by this vaccination regimen. Part of its efficacy certainly relies on the generation and maintenance of large numbers of specific lymphocytes. Other specific characteristics may also be important, and studies on this direction have only recently been initiated. So far, the characterization of these protective, long-lived T cell populations suggests that there is a high frequency of polyfunctional T cells; these cells cover a large breadth and display a T effector memory (TEM) phenotype. These TEM cells are capable of proliferating after an infectious challenge and are highly refractory to apoptosis due to a control of the expression of pro-apoptotic receptors such as CD95. Also, they do not undergo significant long-term immunological erosion. Understanding the mechanisms that control the generation and maintenance of the protective activity of these long-lived TEM cells will certainly provide important insights into the physiology of CD8(+) T cells and pave the way for the design of new or improved vaccines.
Collapse
Affiliation(s)
- José R Vasconcelos
- Centro de Terapia Celular e Molecular, Universidade Federal de São Paulo - Escola Paulista de Medicina São Paulo, São Paulo, Brazil ; Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo - Escola Paulista de Medicina São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
To assess the efficacy of HIV vaccine candidates or preventive treatment, many research groups have started to challenge monkeys repeatedly with low doses of the virus. Such challenge data provide a unique opportunity to assess the importance of exposure history for the acquisition of the infection. I developed stochastic models to analyze previously published challenge data. In the mathematical models, I allowed for variation of the animals' susceptibility to infection across challenge repeats, or across animals. In none of the studies I analyzed, I found evidence for an immunizing effect of non-infecting challenges, and in most studies, there is no evidence for variation in the susceptibilities to the challenges across animals. A notable exception was a challenge experiment by Letvin et al. Sci Translat Med (2011) conducted with the strain SIVsmE660. The challenge data of this experiment showed significant susceptibility variation from animal-to-animal, which is consistent with previously established genetic differences between the involved animals. For the studies which did not show significant immunizing effects and susceptibility differences, I conducted a power analysis and could thus exclude a very strong immunization effect for some of the studies. These findings validate the assumption that non-infecting challenges do not immunize an animal — an assumption that is central in the argument that repeated low-dose challenge experiments increase the statistical power of preclinical HIV vaccine trials. They are also relevant for our understanding of the role of exposure history for HIV acquisition and forecasting the epidemiological spread of HIV. Individuals are exposed to Human Immunodeficiency Virus (HIV) many times before they contract the virus. It is not known what an instance of exposure, which does not result in infection, does to the host. Frequent exposures to the virus are hypothesized to immunize an individual, and result in resistance to infection with HIV. This hypothesis may explain the resistance observed in some individuals despite frequent exposure to the virus. Since it is very difficult to monitor the HIV exposure and infection status of humans, this question is easier to address in animal models. I took data from previously published infection experiments of monkeys with Simian Immunodeficiency Virus (SIV) and analyzed them with newly developed mathematical models. I found that there is no evidence that challenging monkeys with the virus reduces their susceptibility to infection. These findings have important repercussions for the testing of HIV vaccines in monkeys, and also for our understanding of the role of exposure history for the acquisition of HIV.
Collapse
|
15
|
Developing a neonatal HIV vaccine: insights from macaque models of pediatric HIV/AIDS. Curr Opin HIV AIDS 2012; 2:367-74. [PMID: 19372914 DOI: 10.1097/coh.0b013e3282cecf21] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW This review analyzes recent findings from nonhuman primate models of HIV/AIDS that are most relevant to developing active neonatal vaccine strategies against HIV breast milk transmission. We focus on studies published from 2005 to early 2007 that have characterized simian immunodeficiency virus or simian/human immunodeficiency virus transmission and the efficacy of HIV vaccine strategies in neonatal macaques. RECENT FINDINGS Nonhuman primate models of natural HIV breast milk transmission recapitulate many features of infection in human infants; however, the variation in timing and overall low rate of infection in these models precludes their use in conducting vaccine studies. Oral inoculation of infant macaques with defined viral inocula results in reliable transmission and is an efficient model for evaluating neonatal HIV vaccine strategies. All HIV vaccine strategies tested in neonatal macaques are immunogenic, but only a subset of these vaccines confer significant protection against virus acquisition or simian AIDS after oral challenge. SUMMARY Candidate HIV vaccine strategies can elicit virus-specific humoral and cell-mediated immune responses in newborn primates; however, vaccine immunogenicity in infant macaques is not a reliable criterion for predicting a vaccine's efficacy against oral virus challenge exposure.
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW The major target groups for an HIV vaccine include breastfeeding infants and adolescents. Differential immune maturity in these age groups may significantly impact vaccine efficacy, and should be taken into account when developing vaccines. Here we review these differences, with an emphasis on the immune response to vaccines for HIV and other pathogens. Recommendations for potential adaptation of current HIV vaccines are also made. RECENT FINDINGS An effective neonatal vaccine needs to be immunogenic in the presence of maternal antibody, and must induce cytotoxic T-lymphocyte responses, neutralizing antibody responses, both systemic and mucosal. There is renewed hope in the possibility of stimulating neutralizing antibodies with HIV vaccination. DNA vaccines are promising for neonates, but will need appropriate boosting. Certain adjuvants and vector delivery systems are more suitable for neonates. Adolescents may have stronger immune responses to HIV vaccines than adults, and will also require induction of mucosal neutralizing humoral and cellular immunity. SUMMARY Some current HIV vaccine strategies may need adaptation for neonates and suitable product development should be accelerated. Vaccines could induce better responses in adolescents and therefore should not be discarded prematurely. Development of vaccines that have potential for these age groups is an urgent global priority.
Collapse
|
17
|
Heterologous protection elicited by candidate monomeric recombinant HIV-1 gp120 vaccine in the absence of cross neutralising antibodies in a macaque model. Retrovirology 2012; 9:56. [PMID: 22799593 PMCID: PMC3418562 DOI: 10.1186/1742-4690-9-56] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 05/09/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Current data suggest that an efficacious human immunodeficiency virus type 1 (HIV-1) vaccine should elicit both adaptive humoral and cell mediated immune responses. Such a vaccine will also need to protect against infection from a range of heterologous viral variants. Here we have developed a simian-human immunodeficiency virus (SHIV) based model in cynomolgus macaques to investigate the breadth of protection conferred by HIV-1W61D recombinant gp120 vaccination against SHIVsbg and SHIVSF33 challenge, and to identify correlates of protection. RESULTS High titres of anti-envelope antibodies were detected in all vaccinees. The antibodies reacted with both the homologous HIV-1W61D and heterologous HIV-1IIIB envelope rgp120 which has an identical sequence to the SHIVsbg challenge virus. Significant titres of virus neutralising antibodies were detected against SHIVW61D expressing an envelope homologous with the vaccine, but only limited cross neutralisation against SHIVsbg, SHIV-4 and SHIVSF33 was observed. Protection against SHIVsbg infection was observed in vaccinated animals but none was observed against SHIVSF33 challenge. Transfer of immune sera from vaccinated macaques to naive recipients did not confer protection against SHIVsbg challenge. In a follow-up study, T cell proliferative responses detected after immunisation with the same vaccine against a single peptide present in the second conserved region 2 of HIV-1 W61D and HIV-1 IIIB gp120, but not SF33 gp120. CONCLUSIONS Following extended vaccination with a HIV-1 rgp120 vaccine, protection was observed against heterologous virus challenge with SHIVsbg, but not SHIVSF33. Protection did not correlate with serological responses generated by vaccination, but might be associated with T cell proliferative responses against an epitope in the second constant region of HIV-1 gp120. Broader protection may be obtained with recombinant HIV-1 envelope based vaccines formulated with adjuvants that generate proliferative T cell responses in addition to broadly neutralising antibodies.
Collapse
|
18
|
Cox JH, Ferrari MG, Earl P, Lane JR, Jagodzinski LL, Polonis VR, Kuta EG, Boyer JD, Ratto-Kim S, Eller LA, Pham DT, Hart L, Montefiori D, Ferrari G, Parrish S, Weiner DB, Moss B, Kim JH, Birx D, VanCott TC. Inclusion of a CRF01_AE HIV envelope protein boost with a DNA/MVA prime-boost vaccine: Impact on humoral and cellular immunogenicity and viral load reduction after SHIV-E challenge. Vaccine 2012; 30:1830-40. [PMID: 22234262 PMCID: PMC3324265 DOI: 10.1016/j.vaccine.2011.12.131] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 12/21/2011] [Accepted: 12/28/2011] [Indexed: 01/13/2023]
Abstract
The current study assessed the immunogenicity and protective efficacy of various prime-boost vaccine regimens in rhesus macaques using combinations of recombinant DNA (rDNA), recombinant MVA (rMVA), and subunit gp140 protein. The rDNA and rMVA vectors were constructed to express Env from HIV-1 subtype CRF01_AE and Gag-Pol from CRF01_AE or SIVmac 239. One of the rMVAs, MVA/CMDR, has been recently tested in humans. Immunizations were administered at months 0 and 1 (prime) and months 3 and 6 (boost). After priming, HIV env-specific serum IgG was detected in monkeys receiving gp140 alone or rMVA but not in those receiving rDNA. Titers were enhanced in these groups after boosting either with gp140 alone or with rMVA plus gp140. The groups that received the rDNA prime developed env-specific IgG after boosting with rMVA with or without gp140. HIV Env-specific serum IgG binding antibodies were elicited more frequently and of higher titer, and breadth of neutralizing antibodies was increased with the inclusion of the subunit Env boost. T cell responses were measured by tetramer binding to Gag p11c in Mamu-A*01 macaques, and by IFN-γ ELISPOT assay to SIV-Gag. T cell responses were induced after vaccination with the highest responses seen in macaques immunized with rDNA and rMVA. Macaques were challenged intravenously with a novel SHIV-E virus (SIVmac239 Gag-Pol with an HIV-1 subtype E-Env CAR402). Post challenge with SHIV-E, antibody titers were boosted in all groups and peaked at 4 weeks. Robust T cell responses were seen in all groups post challenge and in macaques immunized with rDNA and rMVA a clear boosting of responses was seen. A greater than two-log drop in RNA copies/ml at peak viremia and earlier set point was achieved in macaques primed with rDNA, and boosted with rMVA/SHIV-AE plus gp140. Post challenge viremia in macaques immunized with other regimens was not significantly different to that of controls. These results demonstrate that a gp140 subunit and inclusion of SIV Gag-Pol may be critical for control of SHIV post challenge.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- Antibodies, Neutralizing/blood
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Products, gag/immunology
- Gene Products, pol/immunology
- HIV Antibodies/blood
- HIV-1/immunology
- Immunity, Cellular
- Immunity, Humoral
- Immunization, Secondary
- Immunoglobulin G/blood
- Macaca mulatta
- Male
- Simian Immunodeficiency Virus/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Load
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- env Gene Products, Human Immunodeficiency Virus/immunology
Collapse
|
19
|
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
|
20
|
Molecular evolution analysis of the human immunodeficiency virus type 1 envelope in simian/human immunodeficiency virus-infected macaques: implications for challenge dose selection. J Virol 2011; 85:10332-45. [PMID: 21795341 DOI: 10.1128/jvi.05290-11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Since the demonstration that almost 80% of human immunodeficiency virus type 1 (HIV-1) infections result from the transmission of a single variant from the donor, biological features similar to those of HIV mucosal transmission have been reported for macaques inoculated with simian immunodeficiency virus (SIV). Here we describe the early diversification events and the impact of challenge doses on viral kinetics and on the number of variants transmitted in macaques infected with the chimeric simian/human immunodeficiency virus SHIV(sf162p4). We show that there is a correlation between the dose administered and the number of variants transmitted and that certain inoculum variants are preferentially transmitted. This could provide insight into the viral determinants of transmission and could aid in vaccine development. Challenge through the mucosal route with high doses results in the transmission of multiple variants in all the animals. Such an unrealistic scenario could underestimate potential intervention measures. We thus propose the use of molecular evolution analysis to aid in the determination of challenge doses that better mimic the transmission dynamics seen in natural HIV-1 infection.
Collapse
|
21
|
Goepfert PA, Elizaga ML, Sato A, Qin L, Cardinali M, Hay CM, Hural J, DeRosa SC, DeFawe OD, Tomaras GD, Montefiori DC, Xu Y, Lai L, Kalams SA, Baden LR, Frey SE, Blattner WA, Wyatt LS, Moss B, Robinson HL. Phase 1 safety and immunogenicity testing of DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J Infect Dis 2011; 203:610-9. [PMID: 21282192 DOI: 10.1093/infdis/jiq105] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recombinant DNA and modified vaccinia virus Ankara (rMVA) vaccines represent a promising approach to an HIV/AIDS vaccine. This Phase 1 clinical trial compared the safety and immunogenicity of a rMVA vaccine administered with and without DNA vaccine priming METHODS GeoVax pGA2/JS7 DNA (D) and MVA/HIV62 (M) vaccines encode noninfectious virus-like particles. Intramuscular needle injections were used to deliver placebo, 2 doses of DNA followed by 2 doses of rMVA (DDMM), one dose of DNA followed by 2 doses of rMVA (DMM), or 3 doses of rMVA (MMM) to HIV-seronegative participants. RESULTS Local and systemic symptoms were mild or moderate. Immune response rates for CD4 + and CD8 + T cells were highest in the DDMM group and lowest in the MMM group (77% vs 43% CD4 + and 42% vs 17% CD8 +). In contrast, response rates for Env binding and neutralizing Ab were highest in the MMM group. The DMM group had intermediate response rates. A 1/10th-dose DDMM regimen induced similar T cell but reduced Ab response rates compared with the full-dose DDMM. CONCLUSIONS MVA62 was well tolerated and elicited different patterns of T cell and Ab responses when administered alone or in combination with the JS7 DNA vaccine.
Collapse
Affiliation(s)
- Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, AL 35294, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Phase 1 safety and immunogenicity evaluation of ADMVA, a multigenic, modified vaccinia Ankara-HIV-1 B'/C candidate vaccine. PLoS One 2010; 5:e8816. [PMID: 20111599 PMCID: PMC2810329 DOI: 10.1371/journal.pone.0008816] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Accepted: 11/11/2009] [Indexed: 01/30/2023] Open
Abstract
Background We conducted a Phase I dose-escalation trial of ADMVA, a Clade-B'/C-based HIV-1 candidate vaccine expressing env, gag, pol, nef, and tat in a modified vaccinia Ankara viral vector. Sequences were derived from a prevalent circulating HIV-1 recombinant form in Yunnan, China, an area of high HIV incidence. The objective was to evaluate the safety and immunogenicity of ADMVA in human volunteers. Methodology/Principal Findings ADMVA or placebo was administered intramuscularly at months 0, 1 and 6 to 50 healthy adult volunteers not at high risk for HIV-1. In each dosage group [1×107 (low), 5×107 (mid), or 2.5×108 pfu (high)] volunteers were randomized in a 3∶1 ratio to receive ADMVA or placebo in a double-blinded design. Subjects were followed for local and systemic reactogenicity, adverse events including cardiac adverse events, and clinical laboratory parameters. Study follow up was 18 months. Humoral immunogenicity was evaluated by anti-gp120 binding ELISA, immunoflourescent staining, and HIV-1 neutralization. Cellular immunogenicity was assessed by a validated IFNγ ELISpot assay and intracellular cytokine staining. Anti-vaccinia binding titers were measured by ELISA. ADMVA was generally well-tolerated, with no vaccine-related serious adverse events or cardiac adverse events. Local or systemic reactogenicity events were reported by 77% and 78% of volunteers, respectively. The majority of events were of mild intensity. The IFNγ ELISpot response rate to any HIV antigen was 0/12 (0%) in the placebo group, 3/12 (25%) in the low dosage group, 6/12 (50%) in the mid dosage group, and 8/13 (62%) in the high dosage group. Responses were often multigenic and occasionally persisted up to one year post vaccination. Antibodies to gp120 were detected in 0/12 (0%), 8/13 (62%), 6/12 (50%) and 10/13 (77%) in the placebo, low, mid, and high dosage groups, respectively. Antibodies persisted up to 12 months after vaccination, with a trend toward agreement with the ability to neutralize HIV-1 SF162 in vitro. Two volunteers mounted antibodies that were able to neutralize clade-matched viruses. Conclusions/Significance ADMVA was well-tolerated and elicited durable humoral and cellular immune responses. Trial Registration Clinicaltrials.gov NCT00252148
Collapse
|
23
|
Vasan S, Schlesinger SJ, Huang Y, Hurley A, Lombardo A, Chen Z, Than S, Adesanya P, Bunce C, Boaz M, Boyle R, Sayeed E, Clark L, Dugin D, Schmidt C, Song Y, Seamons L, Dally L, Ho M, Smith C, Markowitz M, Cox J, Gill DK, Gilmour J, Keefer MC, Fast P, Ho DD. Phase 1 safety and immunogenicity evaluation of ADMVA, a multigenic, modified vaccinia Ankara-HIV-1 B'/C candidate vaccine. PLoS One 2010; 5:e8617. [PMID: 20111582 PMCID: PMC2799527 DOI: 10.1371/journal.pone.0008617] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Accepted: 11/11/2009] [Indexed: 01/20/2023] Open
Abstract
Background We conducted a Phase I dose escalation trial of ADVAX, a DNA-based candidate HIV-1 vaccine expressing Clade C/B' env, gag, pol, nef, and tat genes. Sequences were derived from a prevalent circulating recombinant form in Yunnan, China, an area of high HIV-1 incidence. The objective was to evaluate the safety and immunogenicity of ADVAX in human volunteers. Methodology/Principal Findings ADVAX or placebo was administered intramuscularly at months 0, 1 and 3 to 45 healthy volunteers not at high risk for HIV-1. Three dosage levels [0.2 mg (low), 1.0 mg (mid), and 4.0 mg (high)] were tested. Twelve volunteers in each dosage group were assigned to receive ADVAX and three to receive placebo in a double-blind design. Subjects were followed for local and systemic reactogenicity, adverse events, and clinical laboratory parameters. Study follow up was 18 months. Humoral immunogenicity was evaluated by anti-gp120 binding ELISA. Cellular immunogenicity was assessed by a validated IFNγ ELISpot assay and intracellular cytokine staining. ADVAX was safe and well-tolerated, with no vaccine-related serious adverse events. Local and systemic reactogenicity events were reported by 64% and 42% of vaccine recipients, respectively. The majority of events were mild. The IFNγ ELISpot response rates to any HIV antigen were 0/9 (0%) in the placebo group, 3/12 (25%) in the low-dosage group, 4/12 (33%) in the mid-dosage group, and 2/12 (17%) in the high-dosage group. Overall, responses were generally transient and occurred to each gene product, although volunteers responded to single antigens only. Binding antibodies to gp120 were not detected in any volunteers, and HIV seroconversion did not occur. Conclusions/Significance ADVAX delivered intramuscularly is safe, well-tolerated, and elicits modest but transient cellular immune responses. Trial Registration Clinicaltrials.gov NCT00249106
Collapse
Affiliation(s)
- Sandhya Vasan
- Aaron Diamond AIDS Research Center, New York, New York, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Minang JT, Trivett MT, Bolton DL, Trubey CM, Estes JD, Li Y, Smedley J, Pung R, Rosati M, Jalah R, Pavlakis GN, Felber BK, Piatak M, Roederer M, Lifson JD, Ott DE, Ohlen C. Distribution, persistence, and efficacy of adoptively transferred central and effector memory-derived autologous simian immunodeficiency virus-specific CD8+ T cell clones in rhesus macaques during acute infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:315-26. [PMID: 19949091 PMCID: PMC2797560 DOI: 10.4049/jimmunol.0902410] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Plasma viremia decreases coincident with the appearance of virus-specific CD8(+) T cells during acute HIV or SIV infection. This finding, along with demonstrations of viral mutational escape from CD8(+) T cell responses and transient increase in plasma viremia after depletion of CD8(+) T cells in SIV-infected monkeys strongly suggest a role for CD8(+) T cells in controlling HIV/SIV. However, direct quantitative or qualitative correlates between CD8(+) T cell activity and virus control have not been established. To directly assess the impact of large numbers of virus-specific CD8(+) T cells present at time of SIV infection, we transferred in vitro expanded autologous central and effector memory-derived Gag CM9-, Nef YY9-, and Vif WY8-specific CD8(+) T cell clones to acutely infected rhesus macaques. The cells persisted in PBMCs between 4 and 9 d, but were not detected in gut-associated lymphoid tissue or lymph nodes. Interestingly, a high frequency of the infused cells localized to the lungs, where they persisted at high frequency for >6 wk. Although persisting cells in the lungs were Ag reactive, there was no measurable effect on virus load. Sequencing of virus from the animal receiving Nef YY9-specific CD8(+) T cells demonstrated an escape mutation in this epitope <3 wk postinfection, consistent with immune selection pressure by the infused cells. These studies establish methods for adoptive transfer of autologous SIV-specific CD8(+) T cells for evaluating immune control during acute infection and demonstrate that infused cells retain function and persist for at least 2 mo in specific tissues.
Collapse
Affiliation(s)
- Jacob T. Minang
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Matthew T. Trivett
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Diane L. Bolton
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892
| | - Charles M. Trubey
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Yuan Li
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Jeremy Smedley
- Laboratory Animal Science Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Rhonda Pung
- Laboratory Animal Science Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Margherita Rosati
- Human Retrovirus Section, NCI-Frederick, Frederick, Maryland 21702, USA
| | - Rashmi Jalah
- Human Retrovirus Pathogenesis Section, NCI-Frederick, Frederick, Maryland 21702, USA
| | | | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, NCI-Frederick, Frederick, Maryland 21702, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - David E. Ott
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| | - Claes Ohlen
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA
| |
Collapse
|
25
|
Abstract
Evaluation of HIV vaccine candidates in nonhuman primates (NHPs) is a critical step toward developing a successful vaccine to control the HIV pandemic. Historically, HIV vaccine regimens have been tested in NHPs by administering a single high dose of the challenge virus. More recently, evaluation of candidate HIV vaccines has entailed repeated low-dose challenges, which more closely mimic typical exposure in natural transmission settings. In this article, we consider evaluation of the type and magnitude of vaccine efficacy from such experiments. Based on the principal stratification framework, we also address evaluation of potential immunological surrogate endpoints for infection.
Collapse
Affiliation(s)
- Michael G Hudgens
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | |
Collapse
|
26
|
Protease cleavage sites in HIV-1 gp120 recognized by antigen processing enzymes are conserved and located at receptor binding sites. J Virol 2009; 84:1513-26. [PMID: 19939935 DOI: 10.1128/jvi.01765-09] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The identification of vaccine immunogens able to elicit broadly neutralizing antibodies (bNAbs) is a major goal in HIV vaccine research. Although it has been possible to produce recombinant envelope glycoproteins able to adsorb bNAbs from HIV-positive sera, immunization with these proteins has failed to elicit antibody responses effective against clinical isolates of HIV-1. Thus, the epitopes recognized by bNAbs are present on recombinant proteins, but they are not immunogenic. These results led us to consider the possibility that changes in the pattern of antigen processing might alter the immune response to the envelope glycoprotein to better elicit protective immunity. In these studies, we have defined protease cleavage sites on HIV gp120 recognized by three major human proteases (cathepsins L, S, and D) important for antigen processing and presentation. Remarkably, six of the eight sites identified in gp120 were highly conserved and clustered in regions of the molecule associated with receptor binding and/or the binding of neutralizing antibodies. These results suggested that HIV may have evolved to take advantage of major histocompatibility complex (MHC) class II antigen processing enzymes in order to evade or direct the antiviral immune response.
Collapse
|
27
|
Kersh EN, Luo W, Adams DR, Srinivasan P, Smith JM, Promadej-Lanier N, Ellenberger D, Garcia-Lerma JG, Butera S, Otten R. Repeated rectal SHIVSF162P3 exposures do not consistently induce sustained T cell responses prior to systemic infection in the repeat-low dose preclinical macaque model. AIDS Res Hum Retroviruses 2009; 25:905-17. [PMID: 19689194 DOI: 10.1089/aid.2008.0287] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The macaque model of repeated SHIV exposures is increasingly used as a preclinical tool to evaluate biomedical HIV intervention strategies. It is unclear whether multiple virus exposures induce immune responses in macaques, as documented in uninfected individuals repeatedly exposed to HIV. We here address whether repeated, rectal SHIV(SF162P3) exposures lead to systemic T cell activation in 12 rhesus macaques, and whether this is associated with increased infection resistance. Eight macaques became systemically infected after 2-7 exposures, three macaques were less susceptible (infection after 10-12 exposures), and one macaque remained uninfected after 14 exposures. PBMCs were retrospectively monitored for increases in T cell activation by analyzing the proportion of CD8(+) T cells, recently activated or proliferated T cells (markers CD38, Ki67), a marker for cytotoxicity (granzyme B), or T cell-produced plasma cytokines (IFN-gamma, RANTES, IL-2). Repeated virus exposures did not induce sustained, potent, or diverse T cell responses prior to systemic infection. Some changes occurred in the analyzed parameters during repeated virus exposures, but similar T cell activities were also observed in five SHIV-unexposed control macaques. Thus, we found no evidence that delayed infection or resistance to infection was associated with systemic, long-lasting, protective T cell responses to repeated rectal virus exposures. Our results provide further insights into the repeat exposure macaque model. We find that this model can be used for testing biomedical prevention strategies without concern of eliciting a systemic vaccination effect.
Collapse
Affiliation(s)
- Ellen N. Kersh
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Wei Luo
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Debra R. Adams
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Priya Srinivasan
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - James M. Smith
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Nattawan Promadej-Lanier
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Dennis Ellenberger
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - J. Gerardo Garcia-Lerma
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Salvatore Butera
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Ron Otten
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| |
Collapse
|
28
|
Hudgens MG, Gilbert PB, Mascola JR, Wu CD, Barouch DH, Self SG. Power to detect the effects of HIV vaccination in repeated low-dose challenge experiments. J Infect Dis 2009; 200:609-13. [PMID: 19591571 PMCID: PMC2754821 DOI: 10.1086/600891] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Simulation studies were conducted to estimate the statistical power of repeated low-dose challenge experiments performed in nonhuman primates to detect the effect of a candidate human immunodeficiency virus vaccine. The effect of various design parameters on power was explored. Results of simulation studies indicate that repeated low-dose challenge studies with a total sample of size 50 (25 animals/arm) typically provide adequate power to detect a 50% reduction in the per-exposure probability of infection resulting from vaccination. Power generally increases with the maximum number of allowable challenges per animal, the per-exposure risk of infection in control animals, and the proportion of animals susceptible to infection.
Collapse
Affiliation(s)
- Michael G Hudgens
- Department of Biostatistics, School of Public Health, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Promadej-Lanier N, Smith JM, Srinivasan P, McCoy CF, Butera S, Woolfson AD, Malcolm RK, Otten RA. Development and evaluation of a vaginal ring device for sustained delivery of HIV microbicides to non-human primates. J Med Primatol 2009; 38:263-71. [DOI: 10.1111/j.1600-0684.2009.00354.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
The effect of ongoing exposure dynamics in dose response relationships. PLoS Comput Biol 2009; 5:e1000399. [PMID: 19503605 PMCID: PMC2685010 DOI: 10.1371/journal.pcbi.1000399] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Accepted: 05/04/2009] [Indexed: 11/20/2022] Open
Abstract
Characterizing infectivity as a function of pathogen dose is integral to
microbial risk assessment. Dose-response experiments usually administer doses to
subjects at one time. Phenomenological models of the resulting data, such as the
exponential and the Beta-Poisson models, ignore dose timing and assume
independent risks from each pathogen. Real world exposure to pathogens, however,
is a sequence of discrete events where concurrent or prior pathogen arrival
affects the capacity of immune effectors to engage and kill newly arriving
pathogens. We model immune effector and pathogen interactions during the period
before infection becomes established in order to capture the dynamics generating
dose timing effects. Model analysis reveals an inverse relationship between the
time over which exposures accumulate and the risk of infection. Data from one
time dose experiments will thus overestimate per pathogen infection risks of
real world exposures. For instance, fitting our model to one time dosing data
reveals a risk of 0.66 from 313 Cryptosporidium parvum
pathogens. When the temporal exposure window is increased 100-fold using the
same parameters fitted by our model to the one time dose data, the risk of
infection is reduced to 0.09. Confirmation of this risk prediction requires data
from experiments administering doses with different timings. Our model
demonstrates that dose timing could markedly alter the risks generated by
airborne versus fomite transmitted pathogens. We model the relationship between the temporal patterns of pathogen exposure and
infection take off within people. Since different routes of transmission (e.g.,
airborne versus surface transfer routes) may result in different temporal
patterns of exposure, this model helps to better compare the risks of
transmission from one person to another through these different routes. Previous
models assumed that the risk of infection is the same whether pathogens are
inoculated all at once or over one day. Our model, in contrast, captures how one
pathogen affects the potential of immunity to keep concurrently or subsequently
arriving particles from initiating an infection. Since the pattern of timing of
airborne and surface spread pathogen arrivals differ, our model shows that each
airborne pathogen could carry less risk than each surface transmitted pathogen.
Unfortunately, data to fully fit our model are not currently available.
Therefore new experiments will have to be conducted where doses are given across
different temporal windows.
Collapse
|
31
|
Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, Li D, Gilbert PB, Lama JR, Marmor M, Del Rio C, McElrath MJ, Casimiro DR, Gottesdiener KM, Chodakewitz JA, Corey L, Robertson MN, Step Study Protocol Team. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 2008; 372:1881-1893. [PMID: 19012954 PMCID: PMC2721012 DOI: 10.1016/s0140-6736(08)61591-3] [Citation(s) in RCA: 1355] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Observational data and non-human primate challenge studies suggest that cell-mediated immune responses might provide control of HIV replication. The Step Study directly assessed the efficacy of a cell-mediated immunity vaccine to protect against HIV-1 infection or change in early plasma HIV-1 levels. METHODS We undertook a double-blind, phase II, test-of-concept study at 34 sites in North America, the Caribbean, South America, and Australia. We randomly assigned 3000 HIV-1-seronegative participants by computer-generated assignments to receive three injections of MRKAd5 HIV-1 gag/pol/nef vaccine (n=1494) or placebo (n=1506). Randomisation was prestratified by sex, adenovirus type 5 (Ad5) antibody titre at baseline, and study site. Primary objective was a reduction in HIV-1 acquisition rates (tested every 6 months) or a decrease in HIV-1 viral-load setpoint (early plasma HIV-1 RNA measured 3 months after HIV-1 diagnosis). Analyses were per protocol and modified intention to treat. The study was stopped early because it unexpectedly met the prespecified futility boundaries at the first interim analysis. This study is registered with ClinicalTrials.gov, number NCT00095576. FINDINGS In a prespecified interim analysis in participants with baseline Ad5 antibody titre 200 or less, 24 (3%) of 741 vaccine recipients became HIV-1 infected versus 21 (3%) of 762 placebo recipients (hazard ratio [HR] 1.2 [95% CI 0.6-2.2]). All but one infection occurred in men. The corresponding geometric mean plasma HIV-1 RNA was comparable in infected male vaccine and placebo recipients (4.61 vs 4.41 log(10) copies per mL, one tailed p value for potential benefit 0.66). The vaccine elicited interferon-gamma ELISPOT responses in 75% (267) of the 25% random sample of all vaccine recipients (including both low and high Ad5 antibody titres) on whose specimens this testing was done (n=354). In exploratory analyses of all study volunteers, irrespective of baseline Ad5 antibody titre, the HR of HIV-1 infection between vaccine and placebo recipients was higher in Ad5 seropositive men (HR 2.3 [95% CI 1.2-4.3]) and uncircumcised men (3.8 [1.5-9.3]), but was not increased in Ad5 seronegative (1.0 [0.5-1.9]) or circumcised (1.0 [0.6-1.7]) men. INTERPRETATION This cell-mediated immunity vaccine did not prevent HIV-1 infection or reduce early viral level. Mechanisms for insufficient efficacy of the vaccine and the increased HIV-1 infection rates in subgroups of vaccine recipients are being explored.
Collapse
Affiliation(s)
- Susan P Buchbinder
- HIV Research Section, San Francisco Department of Public Health, San Francisco, CA, USA.
| | | | - Ann Duerr
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Daniel W Fitzgerald
- GHESKIO, Port-au-Prince, Haiti; Weill Medical College of Cornell University, New York, NY, USA
| | - Robin Mogg
- Merck Research Laboratories, North Wales, PA, USA
| | - David Li
- Merck Research Laboratories, North Wales, PA, USA
| | | | - Javier R Lama
- Asociacion Civil IMPACTA Salud y Educacion, Miraflores, Lima, Peru
| | | | | | | | | | | | | | - Lawrence Corey
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | |
Collapse
|
32
|
Anton PA, Ibarrondo FJ, Boscardin WJ, Zhou Y, Schwartz EJ, Ng HL, Hausner MA, Shih R, Elliott J, Hultin PM, Hultin LE, Price C, Fuerst M, Adler A, Wong JT, Yang OO, Jamieson BD. Differential immunogenicity of vaccinia and HIV-1 components of a human recombinant vaccine in mucosal and blood compartments. Vaccine 2008; 26:4617-23. [PMID: 18621451 PMCID: PMC2570228 DOI: 10.1016/j.vaccine.2008.05.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2007] [Revised: 05/07/2008] [Accepted: 05/14/2008] [Indexed: 12/16/2022]
Abstract
Mucosal immune responses induced by HIV-1 vaccines are likely critical for prevention. We report a Phase 1 safety and immunogenicity trial in eight participants using the vaccinia-based TBC-3B vaccine given subcutaneously to determine the relationship between HIV-1 specific systemic and gastrointestinal mucosal responses. Across all subjects, detectable levels of blood vaccinia- and HIV-1-specific antibodies were elicited but none were seen mucosally. While the vaccinia component was immunogenic for CD8(+) T lymphocyte (CTL) responses in both blood and mucosa, it was greater in blood. The HIV-1 component of the vaccine was poorly immunogenic in both blood and mucosa. Although only eight volunteers were studied intensively, the discordance between mucosal and blood responses may highlight mechanisms contributing to recent vaccine failures.
Collapse
Affiliation(s)
- Peter A Anton
- Center for Prevention Research and the UCLA AIDS Institute, David Geffen School of Medicine at UCLA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Aidoo M, Sawadogo S, Bile EC, Yang C, Nkengasong JN, McNicholl JM. Viral, HLA and T cell elements in cross-reactive immune responses to HIV-1 subtype A, CRF01_AE and CRF02_AG vaccine sequence in Ivorian blood donors. Vaccine 2008; 26:4830-9. [PMID: 18640166 DOI: 10.1016/j.vaccine.2008.06.097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 06/21/2008] [Accepted: 06/30/2008] [Indexed: 10/21/2022]
Abstract
Comprehensive understanding of the determinants of cross-subtype immune responses in HIV infection is critical to developing efficacious HIV vaccines against multiple viral subtypes. Because HIV-1 subtype A or recombinants comprising subtype A are prevalent in Africa and parts of Asia where HIV is spreading, we assessed the determinants of cross-subtype immune responses in HIV-infected blood donors from Cote d'Ivoire to peptides from a candidate CRF02_AG vaccine sequence, a subtype A sequence from western Kenya and a CRF01_AE sequence from Thailand. We present evidence that immune recognition of multiple viral subtypes is maintained by recognition of multiple epitopes. Our data suggest that complete escape of HIV from immune recognition is uncommon. Evaluation of these frequently generated cross-reactive responses should be included in immunogenicity trials of HIV vaccines.
Collapse
Affiliation(s)
- M Aidoo
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Systemic and mucosal immunological responses during repeated mucosal SHIV162P3 challenges prior to and following infection in pigtailed macaques. Virology 2008; 375:492-503. [DOI: 10.1016/j.virol.2008.01.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 12/23/2007] [Accepted: 01/28/2008] [Indexed: 11/18/2022]
|
35
|
Juompan LY, Hutchinson K, Montefiori DC, Nidtha S, Villinger F, Novembre FJ. Analysis of the immune responses in chimpanzees infected with HIV type 1 isolates. AIDS Res Hum Retroviruses 2008; 24:573-86. [PMID: 18426337 DOI: 10.1089/aid.2007.0182] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mechanisms of resistance to AIDS development in HIV-1-infected chimpanzees have remained elusive. Unique among chimpanzees naturally or experimentally infected with HIV, several animals of the Yerkes cohort have progressed to clinical AIDS with selection of isolates showing increased pathogenicity for chimpanzees. We compared progressors vs. nonprogressors among the HIV-infected chimpanzees that made up this cohort, eight of which have been infected with HIV-1 for over 14 years. The additional two progressors were infected de novo with chimpanzee-pathogenic HIV, rapidly leading to a progressor status. Nonprogressors were characterized by normal CD4(+) T cell counts and the absence of detectable viremia. In contrast, progressor chimpanzees had relatively high plasma viral loads associated with a dramatic loss of CD4(+) T cells. The analysis of immune responses showed a similar amplitude and breadth of ELISPOT T cell responses in both groups. HIV-specific proliferative responses were, however, absent in the progressor animals, which also exhibited increased levels of immune activation characterized by elevated levels of the circulating chemokines IP-10 and MCP-1. Of interest was the conservation of potent NK cell activity in all animals, potentially contributing to the extended symptom-free survival of progressor animals. Modest anti-HIV antibody titers were detectable in the nonprogressor group, but these antibodies exhibited good neutralizing activity. In progressors, however, two sets of data were noted: in animals that gradually selected for pathogenic isolates, or that were superinfected, very high neutralizing antibody titers were observed, although none to the pathogenic HIV. In contrast, two animals infected de novo with chimpanzee pathogenic HIV failed to mount an extensive humoral response and both failed to develop neutralizing antibodies to the virus. Taken together, pathogenic HIV infection in chimpanzees is associated with rapid loss of CD4(+) T cells and proliferative responses as well as higher levels of immune activation.
Collapse
Affiliation(s)
- Laure Y. Juompan
- Yerkes National Primate Research Center and Department of Microbiology, Emory University, Atlanta, Georgia 30329
- Division of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Karen Hutchinson
- Special Pathogens Branch, Centers for Disease Control, Atlanta, Georgia 30333
| | | | - Soumya Nidtha
- Yerkes National Primate Research Center and Department of Microbiology, Emory University, Atlanta, Georgia 30329
- TransMed Partners, LLC, San Francisco, California
| | - François Villinger
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia 30329
| | - Francis J. Novembre
- Yerkes National Primate Research Center and Department of Microbiology, Emory University, Atlanta, Georgia 30329
| |
Collapse
|
36
|
Kersh EN, Luo W, Adams DR, Mitchell J, Garcia-Lerma JG, Heneine W, Folks TM, Butera S, Otten RA. Short communication: no evidence of occult SHIV infection as demonstrated by CD8+ cell depletion after chemoprophylaxis-induced protection from mucosal infection in rhesus macaques. AIDS Res Hum Retroviruses 2008; 24:543-6. [PMID: 18370590 DOI: 10.1089/aid.2007.0222] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Preexposure prophylaxis (PrEP) with antiretroviral drugs constitutes a promising strategy for HIV prevention. Potent PrEP regimens with reverse transcriptase inhibitors can prevent detectable SHIV infection in a repeated low-dose macaque model that resembles human transmission, supporting plans to quickly move this approach into human trials. However, the possibility remains that extremely low levels of virus replication could nonetheless occur during PrEP and seed viral reservoirs in tissues. Therefore, seemingly protected macaques may harbor occult virus that may be initially contained by cytotoxic T cells, but could emerge later. To explore this possibility, we studied whether CD8(+) cells suppress viremia in four rhesus macaques apparently protected by daily or intermittent Truvada (FTC and tenofovir) during 14 low-dose, rectal SHIV(SF162P3) challenges and during a subsequent drug washout period. CD8(+) cells were efficiently ablated with antibodies in these and two additional control macaques that were previously infected but had reached undetectable virus set points. During 4 weeks of follow-up, all four macaques remained free of plasma viremia and provirus in blood lymphocytes. In contrast, plasma viremia resurged to 10(6) to 10(7) copies per milliliter within 2 weeks in both control macaques. Thus, these results indicate that the undetectable viremia in the PrEP-protected macaques was not due to CD8(+) cells that were containing a low-level infection. Rather, the PrEP treatment created conditions in which infection was prevented, eliminated, or controlled by unknown mechanisms. These data provide important information for PrEP usage to prevent HIV transmission, and fully support the continued pursuit of PrEP prevention measures in humans.
Collapse
Affiliation(s)
- Ellen N. Kersh
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Wei Luo
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Debra R. Adams
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - James Mitchell
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - J. Gerardo Garcia-Lerma
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Walid Heneine
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Thomas M. Folks
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Sal Butera
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Ron A. Otten
- Division of HIV/AIDS prevention, NCHHSTP, CCID, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| |
Collapse
|
37
|
Aidoo M, Otten RA, Rodriguez V, Sariol CA, Martinez M, Kraiselburd E, Robinson H, Folks T, Butera S, Ellenberger D. Absence of SHIV infection in gut and lymph node tissues in rhesus monkeys after repeated rectal challenges following HIV-1 DNA/MVA immunizations. Vaccine 2007; 25:6474-81. [PMID: 17688978 DOI: 10.1016/j.vaccine.2007.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Accepted: 06/11/2007] [Indexed: 10/23/2022]
Abstract
We reported previously the absence of systemic infection in a subset of macaques vaccinated with an HIV-1 DNA/MVA vaccine after 18 or more rectal challenges with low (physiologically relevant) doses of SHIV162P3. To further study the apparent protection, we looked for sequestered virus in gut tissues, lymph nodes, spleen, and testes obtained at necropsy using virus co-culture and nested PCR for SIV Gag, Pol and LTR. There was no evidence of sequestered virus in tissues obtained from the four protected macaques. In contrast, at least one tissue from each of 11 infected animals scored positive by one of these sensitive procedures. Activated PBMC from the protected macaques were not inherently resistant to in vitro infection by the challenge virus. These findings demonstrate that some vaccinated macaques appeared to be free from the challenge virus. Therefore, such T cell-based vaccines may provide some protection when challenge virus doses approach physiological equivalencies.
Collapse
Affiliation(s)
- Michael Aidoo
- Centers for Disease Control and Prevention, 1600 Clifton Road, Mail Stop A-25, Atlanta, GA 30333, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- Margaret I Johnston
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | | |
Collapse
|
39
|
Heeney JL, Plotkin SA. Immunological correlates of protection from HIV infection and disease. Nat Immunol 2007; 7:1281-4. [PMID: 17110946 DOI: 10.1038/ni1206-1281] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The recent meeting on "Immune Correlates of Protection from HIV Infection and Disease" examined new data from a variety of preclinical and clinical settings. These new insights may facilitate vaccine design and clinical evaluation.
Collapse
Affiliation(s)
- Jonathan L Heeney
- Department of Virology, Biomedical Primate Research Centre, University of Leiden, The Netherlands.
| | | |
Collapse
|
40
|
De Boer RJ. Understanding the failure of CD8+ T-cell vaccination against simian/human immunodeficiency virus. J Virol 2007; 81:2838-48. [PMID: 17202215 PMCID: PMC1865966 DOI: 10.1128/jvi.01914-06] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although CD8+ T cells play an important role in controlling viral infections, boosting specific CD8+ T cells by prophylactic vaccination with simian immunodeficiency virus (SIV) epitopes fails to provide sterilizing immunity. Viral replication rates and viral contraction rates after the peak viremia hardly depend on the presence of memory CD8+ T cells. To study these paradoxical findings, we parameterize novel mathematical models for acute SIV and human immunodeficiency virus infection. These models explain that failure of vaccination is due to the fact that effector/target ratios are too low during the viral expansion phase. Because CD8+ T cells require cell-to-cell contacts, immune protection requires high effector/target ratios at the primary site of infection. Effector/target ratios become favorable for immune control at the time of the peak in the viral load when the virus becomes limited by other factors, such as the availability of uninfected target cells. At the viral set point, effector/target ratios are much higher, and perturbations of the number of CD8+ effector cells have a large impact on the viral load. Such protective effector/target ratios are difficult to achieve with nucleic acid- or protein-based vaccines.
Collapse
Affiliation(s)
- Rob J De Boer
- Theoretical Biology UU, Padualaan 8, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
41
|
Affiliation(s)
- Josef Vlasak
- Faculty of Biological Sciences, University of South Bohemia, Ceske Budejovice, Czech Republic
| | | |
Collapse
|