488 results on '"Lifson, A"'
Search Results
2. Electron Tomography Analysis of Envelope Glycoprotein Trimers on HIV and Simian Immunodeficiency Virus Virions
- Author
-
Zhu, Ping, Chertova, Elena, Bess,, Julian, Lifson, Jeffrey D., Arthur, Larry O., Liu, Jun, Taylor, Kenneth A., and Roux, Kenneth H.
- Published
- 2003
3. Myeloid cell tropism enables MHC-E-restricted CD8(+) T cell priming and vaccine efficacy by the RhCMV/SIV vaccine
- Author
-
Scott G. Hansen, Meaghan H. Hancock, Daniel Malouli, Emily E. Marshall, Colette M. Hughes, Kurt T. Randall, David Morrow, Julia C. Ford, Roxanne M. Gilbride, Andrea N. Selseth, Renee Espinosa Trethewy, Lindsey M. Bishop, Kelli Oswald, Rebecca Shoemaker, Brian Berkemeier, William J. Bosche, Michael Hull, Lorna Silipino, Michael Nekorchuk, Kathleen Busman-Sahay, Jacob D. Estes, Michael K. Axthelm, Jeremy Smedley, Danica Shao, Paul T. Edlefsen, Jeffrey D. Lifson, Klaus Früh, Jay A. Nelson, and Louis J. Picker
- Subjects
Immunology ,SAIDS Vaccines ,Simian Acquired Immunodeficiency Syndrome ,Cytomegalovirus ,Vaccine Efficacy ,General Medicine ,CD8-Positive T-Lymphocytes ,Macaca mulatta ,Tropism ,Article ,Major Histocompatibility Complex ,Cytomegalovirus Vaccines ,Epitopes ,MicroRNAs ,Animals ,Myeloid Cells ,Simian Immunodeficiency Virus - Abstract
The strain 68-1 rhesus cytomegalovirus (RhCMV)–based vaccine for simian immunodeficiency virus (SIV) can stringently protect rhesus macaques (RMs) from SIV challenge by arresting viral replication early in primary infection. This vaccine elicits unconventional SIV-specific CD8 + T cells that recognize epitopes presented by major histocompatibility complex (MHC)–II and MHC-E instead of MHC-Ia. Although RhCMV/SIV vaccines based on strains that only elicit MHC-II– and/or MHC-Ia–restricted CD8 + T cells do not protect against SIV, it remains unclear whether MHC-E–restricted T cells are directly responsible for protection and whether these responses can be separated from the MHC-II–restricted component. Using host microRNA (miR)–mediated vector tropism restriction, we show that the priming of MHC-II and MHC-E epitope–targeted responses depended on vector infection of different nonoverlapping cell types in RMs. Selective inhibition of RhCMV infection in myeloid cells with miR-142–mediated tropism restriction eliminated MHC-E epitope–targeted CD8 + T cell priming, yielding an exclusively MHC-II epitope–targeted response. Inhibition with the endothelial cell–selective miR-126 eliminated MHC-II epitope–targeted CD8 + T cell priming, yielding an exclusively MHC-E epitope–targeted response. Dual miR-142 + miR-126–mediated tropism restriction reverted CD8 + T cell responses back to conventional MHC-Ia epitope targeting. Although the magnitude and differentiation state of these CD8 + T cell responses were generally similar, only the vectors programmed to elicit MHC-E–restricted CD8 + T cell responses provided protection against SIV challenge, directly demonstrating the essential role of these responses in RhCMV/SIV vaccine efficacy.
- Published
- 2022
4. CTLA-4 and PD-1 dual blockade induces SIV reactivation without control of rebound after antiretroviral therapy interruption
- Author
-
Jenna Read, Colin T. King, Maria Pino, Mirko Paiardini, Heather Amrine-Madsen, Michael Nekorchuk, Chi Ngai Chan, Cristin M. Galardi, Hong Wang, James Schawalder, Katharine J. Bar, A. Alicia Koblansky, Colleen S. McGary, Kevin Nguyen, Jacob D. Estes, Sherrie Jean, Samuel L. M. Raines, Shane D. Falcinelli, Jeffrey D. Lifson, Shari Gordon, Luca Micci, Guido Silvestri, Andrew Sanderson, Justin L. Harper, Brian Johns, David Favre, Emily Lindemuth, Kathleen Busman-Sahay, Barbara Cervasi, and David M. Margolis
- Subjects
0301 basic medicine ,biology ,business.industry ,T cell ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,Immune checkpoint ,Blockade ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,CTLA-4 ,030220 oncology & carcinogenesis ,Immunology ,medicine ,biology.protein ,Cytotoxic T cell ,Antibody ,business ,CD8 ,B cell - Abstract
The primary human immunodeficiency virus (HIV) reservoir is composed of resting memory CD4+ T cells, which often express the immune checkpoint receptors programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which limit T cell activation via synergistic mechanisms. Using simian immunodeficiency virus (SIV)-infected, long-term antiretroviral therapy (ART)-treated rhesus macaques, we demonstrate that PD-1, CTLA-4 and dual CTLA-4/PD-1 immune checkpoint blockade using monoclonal antibodies is well tolerated, with evidence of bioactivity in blood and lymph nodes. Dual blockade was remarkably more effective than PD-1 blockade alone in enhancing T cell cycling and differentiation, expanding effector-memory T cells and inducing robust viral reactivation in plasma and peripheral blood mononuclear cells. In lymph nodes, dual CTLA-4/PD-1 blockade, but not PD-1 alone, decreased the total and intact SIV-DNA in CD4+ T cells, and SIV-DNA and SIV-RNA in B cell follicles, a major site of viral persistence during ART. None of the tested interventions enhanced SIV-specific CD8+ T cell responses during ART or viral control after ART interruption. Thus, despite CTLA-4/PD-1 blockade inducing robust latency reversal and reducing total levels of integrated virus, the degree of reservoir clearance was still insufficient to achieve viral control. These results suggest that immune checkpoint blockade regimens targeting PD-1 and/or CTLA-4, if performed in people living with HIV with sustained aviremia, are unlikely to induce HIV remission in the absence of additional interventions.
- Published
- 2020
- Full Text
- View/download PDF
5. The ingenol-based protein kinase C agonist GSK445A is a potent inducer of HIV and SIV RNA transcription
- Author
-
Afam A. Okoye, Rémi Fromentin, Hiroshi Takata, Jessica H. Brehm, Yoshinori Fukazawa, Bryan Randall, Marion Pardons, Vincent Tai, Jun Tang, Jeremy Smedley, Michael Axthelm, Jeffrey D. Lifson, Louis J. Picker, David Favre, Lydie Trautmann, and Nicolas Chomont
- Subjects
CD4(+) T-CELLS ,RNA viruses ,Transcription, Genetic ,REVERSAL ,Physiology ,viruses ,Pathology and Laboratory Medicine ,Memory T cells ,White Blood Cells ,ANTIRETROVIRAL THERAPY ,Immunodeficiency Viruses ,Animal Cells ,ANIMAL-MODEL ,IN-VIVOACTIVATION ,Medicine and Health Sciences ,Biology (General) ,RHESUS MACAQUE RHADINOVIRUS ,Protein Kinase C ,LATENT-VIRUS REACTIVATION ,T Cells ,virus diseases ,Virus Latency ,Body Fluids ,Viral Persistence and Latency ,PROSTRATIN ,Blood ,SIV ,Medical Microbiology ,Viral Pathogens ,Viruses ,RNA, Viral ,Simian Immunodeficiency Virus ,Diterpenes ,Cellular Types ,Pathogens ,Anatomy ,Research Article ,QH301-705.5 ,Immune Cells ,Immunology ,Cytotoxic T cells ,Microbiology ,Blood Plasma ,Virology ,Retroviruses ,Genetics ,Animals ,Humans ,Microbial Pathogens ,Molecular Biology ,Blood Cells ,Lentivirus ,Organisms ,HIV ,Biology and Life Sciences ,Cell Biology ,RC581-607 ,Macaca mulatta ,Viral Replication ,INFECTED INDIVIDUALS ,Virus Activation ,Parasitology ,Immunologic diseases. Allergy - Abstract
Activation of the NF-κB signaling pathway by Protein Kinase C (PKC) agonists is a potent mechanism for human immunodeficiency virus (HIV) latency disruption in vitro. However, significant toxicity risks and the lack of evidence supporting their activity in vivo have limited further evaluation of PKC agonists as HIV latency-reversing agents (LRA) in cure strategies. Here we evaluated whether GSK445A, a stabilized ingenol-B derivative, can induce HIV/simian immunodeficiency virus (SIV) transcription and virus production in vitro and demonstrate pharmacological activity in nonhuman primates (NHP). CD4+ T cells from people living with HIV and from SIV+ rhesus macaques (RM) on antiretroviral therapy (ART) exposed in vitro to 25 nM of GSK445A produced cell-associated viral transcripts as well as viral particles at levels similar to those induced by PMA/Ionomycin, indicating that GSK445A can potently reverse HIV/SIV latency. Importantly, these concentrations of GSK445A did not impair the proliferation or survival of HIV-specific CD8+ T cells, but instead, increased their numbers and enhanced IFN-γ production in response to HIV peptides. In vivo, GSK445A tolerability was established in SIV-naïve RM at 15 μg/kg although tolerability was reduced in SIV-infected RM on ART. Increases in plasma viremia following GSK445A administration were suggestive of increased SIV transcription in vivo. Collectively, these results indicate that GSK445A is a potent HIV/SIV LRA in vitro and has a tolerable safety profile amenable for further evaluation in vivo in NHP models of HIV cure/remission., Author summary Antiretroviral therapy (ART) is not a definitive cure for HIV infection, in part, because the virus is able to integrate its genetic material in the host cell and remain in a dormant but fully replication-competent form during ART. These latently-infected cells can persist for long periods of time and remain hidden from the host’s immune system. If ART is stopped, the virus can reactivate from this pool of infected cells and resume HIV replication and disease progression. As such, finding and eliminating cells with latent HIV infection is priority for HIV cure research. One approach is to use compounds referred to as latency-reversing agents, that can induce HIV reactivation during ART. The goal of this approach is to facilitate elimination of infected cells by the virus itself once it reactivates or by the host’s immune system, once virus induction renders the cells detectable by the immune system, while also preventing the virus from infecting new cells due to the continued presence of ART. In this study we report on the activity of a novel latency-reversing agent called GSK445A, a potent activator of the enzyme protein kinase C (PKC). We show that GSK445A can induce HIV and simian immunodeficiency virus (SIV) latency reversal in vitro and has a tolerable saftey profile in nonhuman primates that should permit further testing of this PKC-agonist in strategies to cure HIV.
- Published
- 2022
- Full Text
- View/download PDF
6. OP 2.4 – 00145 No Evidence of Ongoing Viral Replication in SIV-Infected Macaques on Combination Antiretroviral Therapy Initiated in the Chronic Phase of Infection Despite Elevated Residual Plasma Viral Loads
- Author
-
G.Q. Del Prete, M. Nag, T. Immonen, C. Fennessey, W. Bosch, A. Conchas, A.E. Swanstrom, J. Lifson, B.F. Keele, A. Macairan, K. Oswald, R. Fast, R. Shoemaker, L. Silipino, M. Hull, D. Donohue, T. Malys, G. Muthua, M. Breed, and J. Kramer
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health - Published
- 2022
- Full Text
- View/download PDF
7. OP 6.6 – 00134 Viral Suppression in SHIV-infected Rhesus Macaques following AAVmediated Delivery of Closer-to-germline Monoclonal Antibodies
- Author
-
J. Martinez-Navio, S.P. Fuchs, D.E. Mendes, C.P. Ramos Muniz, E.G. Rakasz, G. Gao, J.D. Lifson, and R.C. Desrosiers
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health - Published
- 2022
- Full Text
- View/download PDF
8. PP 2.15 – 00169 Macrophages are the primary source of virus in semen and male genital tract organs in acutely and chronically infected rhesus macaques
- Author
-
C. Deleage, C. Fennessey, J. Harper, S. Florea, L. Lipkey, R. Fast, M. Paiardini, J. Lifson, and B. Keele
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health - Published
- 2022
- Full Text
- View/download PDF
9. OP 2.5 – 00048 Targeting the SIV reservoir with Alemtuzumab
- Author
-
B. Varco-Merth, M. Chaunzwa, D. Duell, A. Marenco, S. Docken, J. Smedley, M.K. Axthelm, S.G. Hansen, M.P. Davenport, J.D. Estes, B. Keele, J.D. Lifson, S.R. Lewin, A.A. Okoye, and L.J. Picker
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health - Published
- 2022
- Full Text
- View/download PDF
10. PP 8.1 – 00003 Optimization of the 5′ cap and untranslated regions enhances the immunogenicity of an mRNA-based therapeutic vaccine in SIV-infected rhesus macaques on ART
- Author
-
W. Omange, B. Varco-Merth, A. Morenco, M.T. Chaunzwa, C. Nkoy, D. Duell, O. Fadeyi, M. Medina, S. Hoffmeister, W. Goodwin, R. Butler, Z. Etaki, H. Park, J. Smedley, M.K. Axhelm, S. Hansen, J. Lifson, J. Gergen, S. Rauch, B. Petsch, L.J. Picker, and A.A. Okoye
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health - Published
- 2022
- Full Text
- View/download PDF
11. OP 8.4 – 00165 TLR agonist and SIV mAbs administered to SIV-infected ART-suppressed macaques did not delay rebound after treatment interruption
- Author
-
H. King, D. Brammer, C. Lehman, M. Roederer, D. Bolton, R. Mason, K. Song, K. Foulds, J. Lifson, P. Darrah, and R. Geleziunas
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health - Published
- 2022
- Full Text
- View/download PDF
12. PP 4.1 – 00013 Vaccine-mediated induction of elite control-associated CD8+ cytotoxic T lymphocytes in Mamu-B*08+ Indian rhesus macaques does not protect against intrarectal SIVmac239 acquisition
- Author
-
B.C. Rosen, M.J. Ricciardi, Nuria Pedreño-Lopez, T.B. Voigt, F.D. Laurino, J.J. Louw, A. Yrizarry-Medina, C. Panayiotou, T. Newbolt, K.L. Weisgrau, J.D. Lifson, R.C. Desrosiers, E.G. Rakasz, and D.I. Watkins
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health - Published
- 2022
- Full Text
- View/download PDF
13. Concordance of immunological events between intrarectal and intravenous SHIVAD8-EO infection when assessed by Fiebig-equivalent staging
- Author
-
Amarendra Pegu, Giulia Fabozzi, Joana Dias, John R. Mascola, Wanwisa Promsote, Cassandra G. Almasri, Richard A. Koup, Lucio Gama, Constantinos Petrovas, John Paul Todd, Jonathan Fintzi, Yoshiaki Nishimura, Mangaiarkarasi Asokan, Kylie March, Malcolm A. Martin, and Jeffrey D. Lifson
- Subjects
Male ,Time Factors ,T cell ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,Viremia ,CD8-Positive T-Lymphocytes ,Serology ,Translational Research, Biomedical ,Administration, Rectal ,Follicular phase ,medicine ,Animals ,Humans ,CXCL13 ,Lymph node ,business.industry ,General Medicine ,Viral Load ,medicine.disease ,Acquired immune system ,Macaca mulatta ,medicine.anatomical_structure ,Immunology ,Disease Progression ,HIV-1 ,Administration, Intravenous ,Female ,Simian Immunodeficiency Virus ,business ,CD8 ,Research Article - Abstract
Primary HIV-1 infection can be classified into six Fiebig stages based on virological and serological laboratory testing, whereas simian-HIV (SHIV) infection in nonhuman primates (NHPs) is defined in time post-infection, making it difficult to extrapolate NHP experiments to the clinics. We identified and extensively characterized the Fiebig-equivalent stages in NHPs challenged intrarectally or intravenously with SHIV(AD8-EO). During the first month post-challenge, intrarectally challenged monkeys were up to 1 week delayed in progression through stages. However, regardless of the challenge route, stages I–II predominated before, and stages V–VI predominated after, peak viremia. Decrease in lymph node (LN) CD4(+) T cell frequency and rise in CD8(+) T cells occurred at stage V. LN virus-specific CD8(+) T cell responses, dominated by degranulation and TNF, were first detected at stage V and increased at stage VI. A similar late elevation in follicular CXCR5(+) CD8(+) T cells occurred, consistent with higher plasma CXCL13 levels at these stages. LN SHIV(AD8-EO) RNA(+) cells were present at stage II, but appeared to decline at stage VI when virions accumulated in follicles. Fiebig-equivalent staging of SHIV(AD8-EO) infection revealed concordance of immunological events between intrarectal and intravenous infection despite different infection progressions, and can inform comparisons of NHP studies with clinical data.
- Published
- 2021
- Full Text
- View/download PDF
14. Interleukin-15 response signature predicts RhCMV/SIV vaccine efficacy
- Author
-
Taryn Urion, Connor B. Driscoll, Rebecca Shoemaker, Emily Ainslie, Rafick Pierre Sekaly, Slim Fourati, Yoshinori Fukazawa, Lynn Law, Richard Green, Fredrik Barrenäs, Ewelina Kosmider, Randy Fast, David Morrow, Paul T. Edlefsen, Kurt T. Randall, Michael K. Axthelm, Andrea N. Selseth, William J. Bosche, Julia C. Ford, Jeffrey D. Lifson, Jan Komorowski, Barbara K. Felber, Jean Chang, Elise Smith, Leanne S. Whitmore, Bryan E. Randall, Danica Shao, Kelli Oswald, Michael Gale, Colette M. Hughes, Inah Golez, George N. Pavlakis, Scott G. Hansen, Daniel J. Newhouse, Xinxia Peng, Louis J. Picker, Wenjun Song, and Roxanne M. Gilbride
- Subjects
0301 basic medicine ,Male ,RNA viruses ,Simian Acquired Immunodeficiency Syndrome ,Cytomegalovirus ,Gene Expression ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Pathology and Laboratory Medicine ,White Blood Cells ,0302 clinical medicine ,Medical Conditions ,Immunodeficiency Viruses ,Animal Cells ,Medicine and Health Sciences ,Cytotoxic T cell ,Public and Occupational Health ,Biology (General) ,Interleukin-15 ,Vaccines ,T Cells ,Viral Vaccine ,SAIDS Vaccines ,Vaccination and Immunization ,Vaccination ,medicine.anatomical_structure ,Infectious Diseases ,SIV ,Interleukin 15 ,Medical Microbiology ,Viral Pathogens ,Immunologi ,Viruses ,Female ,Simian Immunodeficiency Virus ,Pathogens ,Cellular Types ,Research Article ,Infectious Disease Control ,QH301-705.5 ,T cell ,Immune Cells ,Genetic Vectors ,Immunology ,Cytotoxic T cells ,Biology ,Microbiology ,03 medical and health sciences ,Immune system ,Virology ,Retroviruses ,medicine ,Genetics ,Animals ,Molecular Biology ,Microbial Pathogens ,Blood Cells ,Viral vaccines ,Lentivirus ,Organisms ,HIV vaccines ,Biology and Life Sciences ,Cell Biology ,Simian immunodeficiency virus ,RC581-607 ,Vaccine efficacy ,Macaca mulatta ,030104 developmental biology ,Parasitology ,Preventive Medicine ,Immunologic diseases. Allergy ,030215 immunology - Abstract
Simian immunodeficiency virus (SIV) challenge of rhesus macaques (RMs) vaccinated with strain 68–1 Rhesus Cytomegalovirus (RhCMV) vectors expressing SIV proteins (RhCMV/SIV) results in a binary outcome: stringent control and subsequent clearance of highly pathogenic SIV in ~55% of vaccinated RMs with no protection in the remaining 45%. Although previous work indicates that unconventionally restricted, SIV-specific, effector-memory (EM)-biased CD8+ T cell responses are necessary for efficacy, the magnitude of these responses does not predict efficacy, and the basis of protection vs. non-protection in 68–1 RhCMV/SIV vector-vaccinated RMs has not been elucidated. Here, we report that 68–1 RhCMV/SIV vector administration strikingly alters the whole blood transcriptome of vaccinated RMs, with the sustained induction of specific immune-related pathways, including immune cell, toll-like receptor (TLR), inflammasome/cell death, and interleukin-15 (IL-15) signaling, significantly correlating with subsequent vaccine efficacy. Treatment of a separate RM cohort with IL-15 confirmed the central involvement of this cytokine in the protection signature, linking the major innate and adaptive immune gene expression networks that correlate with RhCMV/SIV vaccine efficacy. This change-from-baseline IL-15 response signature was also demonstrated to significantly correlate with vaccine efficacy in an independent validation cohort of vaccinated and challenged RMs. The differential IL-15 gene set response to vaccination strongly correlated with the pre-vaccination activity of this pathway, with reduced baseline expression of IL-15 response genes significantly correlating with higher vaccine-induced induction of IL-15 signaling and subsequent vaccine protection, suggesting that a robust de novo vaccine-induced IL-15 signaling response is needed to program vaccine efficacy. Thus, the RhCMV/SIV vaccine imparts a coordinated and persistent induction of innate and adaptive immune pathways featuring IL-15, a known regulator of CD8+ T cell function, that support the ability of vaccine-elicited unconventionally restricted CD8+ T cells to mediate protection against SIV challenge., Author summary SIV insert-expressing vaccine vectors based on strain 68–1 RhCMV elicit robust, highly effector-memory-biased, unconventionally restricted T cell responses that are associated with an unprecedented level of SIV control after challenge (replication arrest leading to clearance) in slightly over half of vaccinated monkeys. Since efficacy among monkeys vaccinated with the effective 68–1 vaccine is not predicted by standard measures of immunogenicity, we used functional genomics analysis of RhCMV/SIV vaccinated monkeys with known challenge outcomes to identify immune correlates of protection. We found that vaccine efficacy significantly correlates with a vaccine-induced response to IL-15 that includes modulation of immune cell, inflammation, TLR signaling, and cell death programming response pathways. These data suggest that RhCMV/SIV efficacy results from a coordinated and sustained vaccine-mediated induction of innate and adaptive immune pathways featuring IL-15, a known regulator of CD8+ effector-memory T cell function, that cooperates with vaccine-elicited CD8+ T cells to mediate efficacy.
- Published
- 2021
15. Role of IL-15 Signaling in the Pathogenesis of Simian Immunodeficiency Virus Infection in Rhesus Macaques
- Author
-
Rebecca L. Skalsky, Scott W. Wong, Derick M. Duell, Lina Gao, Richard Lum, Michael K. Axthelm, Audrie L. Konfe, Maren Q. DeGottardi, Afam A. Okoye, Devin N. Fachko, Mukta Vaidya, He Li, Byung Park, Louis J. Picker, Yoshinori Fukazawa, Chike O. Abana, Jeffrey D. Lifson, and Anne D. Lewis
- Subjects
Male ,T cell ,Immunology ,Cell ,Simian Acquired Immunodeficiency Syndrome ,Biology ,Article ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Immunology and Allergy ,Rhadinovirus ,Interleukin-15 ,Effector ,virus diseases ,biology.organism_classification ,Macaca mulatta ,Chronic infection ,medicine.anatomical_structure ,Interleukin 15 ,Female ,Simian Immunodeficiency Virus ,CD8 ,Signal Transduction ,030215 immunology - Abstract
Although IL-15 has been implicated in the pathogenic hyperimmune activation that drives progressive HIV and SIV infection, as well as in the generation of HIV/SIV target cells, it also supports NK and T cell homeostasis and effector activity, potentially benefiting the host. To understand the role of IL-15 in SIV infection and pathogenesis, we treated two cohorts of SIVmac239-infected rhesus macaques (RM; Macaca mulatta), one with chronic infection, the other with primary infection, with a rhesusized, IL-15–neutralizing mAb (versus an IgG isotype control) for up to 10 wk (n = 7–9 RM per group). In both cohorts, anti–IL-15 was highly efficient at blocking IL-15 signaling in vivo, causing 1) profound depletion of NK cells in blood and tissues throughout the treatment period; 2) substantial, albeit transient, depletion of CD8+ effector memory T cells (TEM) (but not the naive and central memory subsets); and 3) CD4+ and CD8+ TEM hyperproliferation. In primary infection, reduced frequencies of SIV-specific effector T cells in an extralymphoid tissue site were also observed. Despite these effects, the kinetics and extent of SIV replication, CD4+ T cell depletion, and the onset of AIDS were comparable between anti–IL-15– and control-treated groups in both cohorts. However, RM treated with anti–IL-15 during primary infection manifested accelerated reactivation of RM rhadinovirus. Thus, IL-15 support of NK cell and TEM homeostasis does not play a demonstrable, nonredundant role in SIV replication or CD4+ T cell deletion dynamics but may contribute to immune control of oncogenic γ-herpesviruses.
- Published
- 2019
- Full Text
- View/download PDF
16. Recombinant Herpesvirus Vectors: Durable Immune Responses and Durable Protection against Simian Immunodeficiency Virus SIVmac239 Acquisition
- Author
-
Jeffrey D. Lifson, Lucas Gonzalez-Nieto, Ronald C. Desrosiers, Isabelle M. Castro, Mauricio A. Martins, Eva G. Rakasz, Michael J. Ricciardi, and David I. Watkins
- Subjects
Male ,Rhadinovirus ,Time Factors ,T-Lymphocytes ,viruses ,Genetic Vectors ,Immunology ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Virus ,03 medical and health sciences ,Immunogenicity, Vaccine ,Immunity ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,HIV vaccine ,030304 developmental biology ,0303 health sciences ,030306 microbiology ,SAIDS Vaccines ,virus diseases ,Simian immunodeficiency virus ,biology.organism_classification ,Macaca mulatta ,Vaccination ,HIV Antigens ,Insect Science ,Female ,Simian Immunodeficiency Virus ,Immunologic Memory ,Viral load ,Follow-Up Studies - Abstract
A prophylactic vaccine that confers durable protection against human immunodeficiency virus (HIV) would provide a valuable tool to prevent new HIV/AIDS cases. As herpesviruses establish lifelong infections that remain largely subclinical, the use of persistent herpesvirus vectors to deliver HIV antigens may facilitate the induction of long-term anti-HIV immunity. We previously developed recombinant (r) forms of the gamma-herpesvirus rhesus monkey rhadinovirus (rRRV) expressing a replication-incompetent, near-full-length simian immunodeficiency virus (SIVnfl) genome. We recently showed that 8/16 rhesus macaques (RMs) vaccinated with a rDNA/rRRV-SIVnfl regimen were significantly protected against intrarectal (i.r.) challenge with SIVmac239. Here we investigated the longevity of this vaccine-mediated protection. Despite receiving no additional booster immunizations, the protected rDNA/rRRV-SIVnfl vaccinees maintained detectable cellular and humoral anti-SIV immune responses for more than 1.5 years after the rRRV boost. To assess if these responses were still protective, the rDNA/rRRV-SIVnfl vaccinees were subjected to a second round of marginal-dose i.r. SIVmac239 challenges, with eight SIV-naive RMs serving as concurrent controls. After three SIV exposures, 8/8 control animals became infected, compared to 3/8 vaccinees. This difference in SIV acquisition was statistically significant (P = 0.0035). The three vaccinated monkeys that became infected exhibited significantly lower viral loads than those in unvaccinated controls. Collectively, these data illustrate the ability of rDNA/rRRV-SIVnfl vaccination to provide long-term immunity against stringent mucosal challenges with SIVmac239. Future work is needed to identify the critical components of this vaccine-mediated protection and the extent to which it can tolerate sequence mismatches in the challenge virus. IMPORTANCE We report on the long-term follow-up of a group of rhesus macaques (RMs) that received an AIDS vaccine regimen and were subsequently protected against rectal acquisition of simian immunodeficiency virus (SIV) infection. The vaccination regimen employed included a live recombinant herpesvirus vector that establishes persistent infection in RMs. Consistent with the recurrent SIV antigen expression afforded by this herpesvirus vector, vaccinees maintained detectable SIV-specific immune responses for more than 1.5 years after the last vaccination. Importantly, these vaccinated RMs were significantly protected against a second round of rectal SIV exposures performed 1 year after the first SIV challenge phase. These results are relevant for HIV vaccine development because they show the potential of herpesvirus-based vectors to maintain functional antiretroviral immunity without the need for repeated boosting.
- Published
- 2021
- Full Text
- View/download PDF
17. Evaluating a New Class of AKT/mTOR Activators for HIV Latency-Reversing Activity Ex Vivo and In Vivo
- Author
-
Emilie Besnard, Timothy J. Henrich, Warner C. Greene, Melanie Ott, Jacob D. Estes, William Brantley, Danielle M. Rosenthal, Nevan J. Krogan, Benjamin Varco-Merth, Michael Snape, Satish K. Pillai, Zachary W. Grimmett, Jeffrey D. Lifson, Hannah S. Sperber, Steven G. Deeks, Louise E. Hogan, Louis J. Picker, Michael Nekorchuk, Mark Connors, Stephen A. Migueles, Roland Schwarzer, Bernard Kiernan, Feng Hsiao, Thomas A. Packard, Jeffrey R. Johnson, Philip A. Hull, Nadia R. Roan, Mauricio Montano, Afam A. Okoye, Andrea Gramatica, Eytan Herzig, and Eric Verdin
- Subjects
0303 health sciences ,T cell ,Immunology ,Medizin ,Pharmacology ,Biology ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine.anatomical_structure ,In vivo ,Virology ,Insect Science ,medicine ,Cytotoxic T cell ,Protein kinase B ,030217 neurology & neurosurgery ,PI3K/AKT/mTOR pathway ,CD8 ,Ex vivo ,030304 developmental biology - Abstract
An ability to activate latent HIV-1 expression could benefit many HIV cure strategies, but the first generation of latency reversing agents (LRAs) has proven disappointing. We evaluated AKT/mTOR activators as a potential new class of LRAs. Two glycogen synthase kinase-3 inhibitors (GSK-3i's), SB-216763 and tideglusib (the latter already in phase II clinical trials) that activate AKT/mTOR signaling were tested. These GSK-3i's reactivated latent HIV-1 present in blood samples from aviremic individuals on antiretroviral therapy (ART) in the absence of T cell activation, release of inflammatory cytokines, cell toxicity, or impaired effector function of cytotoxic T lymphocytes or NK cells. However, when administered in vivo to SIV-infected rhesus macaques on suppressive ART, tideglusib exhibited poor pharmacodynamic properties and resulted in no clear evidence of significant SIV latency reversal. Whether alternative pharmacological formulations or combinations of this drug with other classes of LRAs will lead to an effective in vivo latency-reversing strategy remains to be determined.IMPORTANCE If combined with immune therapeutics, latency reversing agents (LRAs) have the potential to reduce the size of the reservoir sufficiently that an engineered immune response can control the virus in the absence of antiretroviral therapy. We have identified a new class of LRAs that do not induce T-cell activation and that are able to potentiate, rather than inhibit, CD8+ T and NK cell cytotoxic effector functions. This new class of LRAs corresponds to inhibitors of glycogen synthase kinase-3. In this work, we have also studied the effects of one member of this drug class, tideglusib, in SIV-infected rhesus monkeys. When tested in vivo, however, tideglusib showed unfavorable pharmacokinetic properties, which resulted in lack of SIV latency reversal. The disconnect between our ex vivo and in vivo results highlights the importance of developing next generation LRAs with pharmacological properties that allow systemic drug delivery in relevant anatomical compartments harboring latent reservoirs.
- Published
- 2021
- Full Text
- View/download PDF
18. CD8 Lymphocyte Depletion Enhances the Latency Reversal Activity of the SMAC Mimetic AZD5582 in ART-Suppressed Simian Immunodeficiency Virus-Infected Rhesus Macaques
- Author
-
Jeffrey D. Lifson, Nils Schoof, Guido Silvestri, Kirk Easley, Ann Chahroudi, Julia Bergild McBrien, Thomas H. Vanderford, Laura E. Liao, Diane G. Carnathan, Deanna A. Kulpa, Mirko Paiardini, Cameron Mattingly, Richard M. Dunham, Alan S. Perelson, Alyssa D. Brooks, David M. Margolis, Ruian Ke, Shan Liang, and Maud Mavigner
- Subjects
biology ,Immunology ,Cell ,Viremia ,medicine.disease ,Microbiology ,Virus ,medicine.anatomical_structure ,Immune system ,In vivo ,Virology ,Insect Science ,biology.protein ,medicine ,Antibody ,Latency (engineering) ,CD8 - Abstract
Inducing latency reversal to reveal infected cells to the host immune system represents a potential strategy to cure HIV infection. In separate studies, we have previously shown that CD8+ T cells may contribute to the maintenance of viral latency and identified a novel SMAC mimetic/IAP inhibitor (AZD5582) capable of reversing HIV/SIV latency in vivo by activating the non-canonical (nc) NF-κB pathway. Here, we use AZD5582 in combination with antibody-mediated depletion of CD8α+ cells to further evaluate the role of CD8+ T cells in viral latency maintenance. Six rhesus macaques (RM) were infected with SIVmac239 and treated with ART starting at week 8 post-infection. After 84-85 weeks of ART, all animals received a single dose of the anti-CD8α depleting antibody (Ab), MT807R1 (50mg/kg, s.c.), followed by 5 weekly doses of AZD5582 (0.1 mg/kg, i.v.). Following CD8α depletion + AZD5582 combined treatment, 100% of RMs experienced on-ART viremia above 60 copies per ml of plasma. In comparator groups of ART-suppressed SIV-infected RMs treated with AZD5582 only or CD8α depletion only, on-ART viremia was experienced by 56% and 57% of the animals respectively. Furthermore, the frequency of increased viremic episodes during the treatment period was greater in the CD8α depletion + AZD5582 group as compared to other groups. Mathematical modeling of virus reactivation suggested that, in addition to viral dynamics during acute infection, CD8α depletion influenced the response to AZD5582. This work suggests that the latency reversal induced by activation of the ncNF-κB signaling pathway with AZD5582 can be enhanced by CD8α+ cell depletion.
- Published
- 2021
- Full Text
- View/download PDF
19. New SHIVs and Improved Design Strategy for Modeling HIV-1 Transmission, Immunopathogenesis, Prevention and Cure
- Author
-
Beatrice H. Hahn, Jessica G. Smith, Yu Ding, Neha Chohan, Brandon F. Keele, Mark G. Lewis, Katharine J. Bar, Alex I. Murphy, Cristian Apetrei, Jeffrey D. Lifson, Hui Li, Fang-Hua Lee, Ivona Pandrea, Thomas N. Denny, Barton F. Haynes, Emily Lindemuth, Juliette Rando, Shuyi Wang, Chengyan Zhao, George M. Shaw, Ryan S. Roark, and Eunlim Kim
- Subjects
Antigenicity ,simian immunodeficiency virus ,viruses ,animal diseases ,Immunology ,Biology ,Gp41 ,Microbiology ,Virus ,Neutralization ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Viral entry ,Virology ,Gene ,Tropism ,030304 developmental biology ,0303 health sciences ,human immunodeficiency virus ,Wild type ,virus diseases ,Vaccine efficacy ,In vitro ,AIDS ,Immunization ,SHIV ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,Antibody ,030217 neurology & neurosurgery - Abstract
SHIV infection of Indian rhesus macaques is an important animal model for studying HIV-1 transmission, prevention, immunopathogenesis, and cure. Such research is timely, given recent progress with active and passive immunization and novel approaches to HIV-1 cure., Previously, we showed that substitution of HIV-1 envelope (Env) residue 375-Ser by bulky aromatic residues enhances binding to rhesus CD4 and enables primary HIV-1 Envs to support efficient replication as simian-human immunodeficiency virus (SHIV) chimeras in rhesus macaques (RMs). Here, we test this design strategy more broadly by constructing SHIVs containing 10 primary Envs corresponding to HIV-1 subtypes A, B, C, AE, and AG. All 10 SHIVs bearing wild-type Env375 residues replicated efficiently in human CD4+ T cells, but only one replicated efficiently in primary rhesus cells. This was a subtype AE SHIV that naturally contained His at Env375. Replacement of wild-type Env375 residues by Trp, Tyr, Phe, or His in the other nine SHIVs led to efficient replication in rhesus CD4+ T cells in vitro and in vivo. Nine SHIVs containing optimized Env375 alleles were grown large-scale in primary rhesus CD4+ T cells to serve as challenge stocks in preclinical prevention trials. These virus stocks were genetically homogeneous, native-like in Env antigenicity and tier 2 neutralization sensitivity, and transmissible by rectal, vaginal, penile, oral, or intravenous routes. To facilitate future SHIV constructions, we engineered a simplified second-generation design scheme and validated it in RMs. Overall, our findings demonstrate that SHIVs bearing primary Envs with bulky aromatic substitutions at Env375 consistently replicate in RMs, recapitulating many features of HIV-1 infection in humans. Such SHIVs are efficiently transmitted by mucosal routes common to HIV-1 infection and can be used to test vaccine efficacy in preclinical monkey trials. IMPORTANCE SHIV infection of Indian rhesus macaques is an important animal model for studying HIV-1 transmission, prevention, immunopathogenesis, and cure. Such research is timely, given recent progress with active and passive immunization and novel approaches to HIV-1 cure. Given the multifaceted roles of HIV-1 Env in cell tropism and virus entry, and as a target for neutralizing and nonneutralizing antibodies, Envs selected for SHIV construction are of paramount importance. Until recently, it has been impossible to strategically design SHIVs bearing clinically relevant Envs that replicate consistently in monkeys. This changed with the discovery that bulky aromatic substitutions at residue Env375 confer enhanced affinity to rhesus CD4. Here, we show that 10 new SHIVs bearing primary HIV-1 Envs with residue 375 substitutions replicated efficiently in RMs and could be transmitted efficiently across rectal, vaginal, penile, and oral mucosa. These findings suggest an expanded role for SHIVs as a model of HIV-1 infection.
- Published
- 2021
20. Non-neutralizing Antibodies May Contribute to Suppression of SIVmac239 Viremia in Indian Rhesus Macaques
- Author
-
Nuria Pedreño-Lopez, Brandon C. Rosen, Walter J. Flores, Matthew J. Gorman, Thomas B. Voigt, Michael J. Ricciardi, Kristin Crosno, Kim L. Weisgrau, Christopher L. Parks, Jeffrey D. Lifson, Galit Alter, Eva G. Rakasz, Diogo M. Magnani, Mauricio A. Martins, and David I. Watkins
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,adoptive transfer ,Genotype ,viruses ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,anti-FcRn Ab ,Viremia ,Receptors, Fc ,medicine.disease_cause ,Antibodies, Viral ,Serology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Immunoadsorption ,antibody depletion ,Original Research ,rhesus macaques (Macaca mulatta) ,biology ,business.industry ,Histocompatibility Antigens Class I ,non-neutralizing antibodies ,virus diseases ,Simian immunodeficiency virus ,Viral Load ,biology.organism_classification ,medicine.disease ,Virology ,Macaca mulatta ,030104 developmental biology ,Viral replication ,SIV ,030220 oncology & carcinogenesis ,Immunoglobulin G ,Lentivirus ,Host-Pathogen Interactions ,biology.protein ,Simian Immunodeficiency Virus ,Antibody ,lcsh:RC581-607 ,business ,Viral load ,Biomarkers ,immunoadsorption - Abstract
The antiviral properties of broadly neutralizing antibodies against HIV are well-documented but no vaccine is currently able to elicit protective titers of these responses in primates. While current vaccine modalities can readily induce non-neutralizing antibodies against simian immunodeficiency virus (SIV) and HIV, the ability of these responses to restrict lentivirus transmission and replication remains controversial. Here, we investigated the antiviral properties of non-neutralizing antibodies in a group of Indian rhesus macaques (RMs) that were vaccinated with vif, rev, tat, nef, and env, as part of a previous study conducted by our group. These animals manifested rapid and durable control of viral replication to below detection limits shortly after SIVmac239 infection. Although these animals had no serological neutralizing activity against SIVmac239 prior to infection, their pre-challenge titers of Env-binding antibodies correlated with control of viral replication. To assess the contribution of anti-Env humoral immune responses to virologic control in two of these animals, we transiently depleted their circulating antibodies via extracorporeal plasma immunoadsorption and inhibition of IgG recycling through antibody-mediated blockade of the neonatal Fc receptor. These procedures reduced Ig serum concentrations by up to 80% and temporarily induced SIVmac239 replication in these animals. Next, we transferred purified total Ig from the rapid controllers into six vaccinated RMs one day before intrarectal challenge with SIVmac239. Although recipients of the hyperimmune anti-SIV Ig fraction were not protected from infection, their peak and chronic phase viral loads were significantly lower than those in concurrent unvaccinated control animals. Together, our results suggest that non-neutralizing Abs may play a role in the suppression of SIVmac239 viremia.
- Published
- 2021
21. A cellular trafficking signal in the SIV envelope protein cytoplasmic domain is strongly selected for in pathogenic infection
- Author
-
Scott P. Lawrence, Samra E. Elser, Workineh Torben, Robert V. Blair, Bapi Pahar, Pyone P. Aye, Faith Schiro, Dawn Szeltner, Lara A. Doyle-Meyers, Beth S. Haggarty, Andrea P. O. Jordan, Josephine Romano, George J. Leslie, Xavier Alvarez, David H. O’Connor, Roger W. Wiseman, Christine M. Fennessey, Yuan Li, Michael Piatak, Jeffrey D. Lifson, Celia C. LaBranche, Andrew A. Lackner, Brandon F. Keele, Nicholas J. Maness, Mark Marsh, and James A. Hoxie
- Subjects
Virology ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Genetics ,Animals ,Gene Products, env ,Simian Immunodeficiency Virus ,Parasitology ,Macaca nemestrina ,Macaca mulatta ,Molecular Biology ,Microbiology ,Endocytosis - Abstract
The HIV/SIV envelope glycoprotein (Env) cytoplasmic domain contains a highly conserved Tyr-based trafficking signal that mediates both clathrin-dependent endocytosis and polarized sorting. Despite extensive analysis, the role of these functions in viral infection and pathogenesis is unclear. An SIV molecular clone (SIVmac239) in which this signal is inactivated by deletion of Gly-720 and Tyr-721 (SIVmac239ΔGY), replicates acutely to high levels in pigtail macaques (PTM) but is rapidly controlled. However, we previously reported that rhesus macaques and PTM can progress to AIDS following SIVmac239ΔGY infection in association with novel amino acid changes in the Env cytoplasmic domain. These included an R722G flanking the ΔGY deletion and a nine nucleotide deletion encoding amino acids 734–736 (ΔQTH) that overlaps therevandtatopen reading frames. We show that molecular clones containing these mutations reconstitute signals for both endocytosis and polarized sorting. In one PTM, a novel genotype was selected that generated a new signal for polarized sorting but not endocytosis. This genotype, together with the ΔGY mutation, was conserved in association with high viral loads for several months when introduced into naïve PTMs. For the first time, our findings reveal strong selection pressure for Env endocytosis and particularly for polarized sorting during pathogenic SIV infectionin vivo.
- Published
- 2022
- Full Text
- View/download PDF
22. RhCMV/SIV tropism modulation programs unconventional CD8+ T cell priming and vaccine efficacy
- Author
-
Meaghan Hancock, Scott Hansen, Daniel Malouli, Emily Marshall, Collette Hughes, Kurt T. Randall, David Morrow, Julia Ford, Roxanne Gilbride, Andrea Selseth, Renee Espinosa Trethewy, Lindsey Bishop, Kelli Oswald, Rebecca Shoemaker, Brian Berkemeier, William Bosche, Michael Hull, Michael Nekorchuk, Kathleen Busman-Sahay, Jacob Estes, Michael Axthelm, Jeremy Smedley, Danica Shao, Paul Edlefsen, Jeffrey Lifson, Klaus Fruh, Jay Nelson, and Louis J Picker
- Subjects
Immunology ,Immunology and Allergy - Abstract
Strain 68-1 rhesus cytomegalovirus (RhCMV) vectors expressing simian immunodeficiency virus (SIV) antigens demonstrate a vaccine efficacy where 50–60% of vaccinated rhesus macaques are protected from SIV challenge. Intriguingly, RhCMV/SIV vectors elicit CD8+ T cells recognizing epitopes presented by MHC-II and MHC-E instead of MHC-Ia. We are studying how these unconventional T cell responses are elicited and contribute to the efficacy against SIV challenge. Here we utilize host microRNA (miRNA)-mediated vector tropism restriction to show that MHC-II- and MHC-E-restricted responses are primed by directly infected, non-overlapping cell types in rhesus macaques. Targeting essential RhCMV genes with myeloid cell-selective miR-142-3p eliminated MHC-E-restricted CD8+ T cell priming, yielding an exclusively MHC-II-restricted response, whereas endothelial cell-selective miR-126-3p targeting eliminated MHC-II-restricted CD8+ T cell priming, yielding an exclusively MHC-E-restricted response. Incorporation of both restriction elements reverts CD8+ T cell responses back to conventional MHC-Ia restriction. Using these otherwise isogenic vectors we show that although they demonstrate similar overall immunogenicity, only the vectors programmed to elicit MHC-E-restricted CD8+ T cell responses provided protection against SIV challenge. The MHC-E-only RhCMV/SIV vaccine efficacy did not exceed that of the parental 68-1 RhCMV/SIV vectors (that elicits both MHC-II and MHC-E responses) indicating that while the MHC-II-restricted CD8+ T cell responses are neutral to overall vaccine efficacy, an additional component of 68-1 RhCMV/SIV-induced immunity contributes to overall vaccine efficacy. This work was supported by the National Institute of Allergy and Infectious Diseases (NIAID) grants UM1 AI124377 and U19 AI128741 to LJP; the Oregon National Primate Research Center Core grant from the National Institutes of Health, Office of the Director (P51 OD011092); contracts from the National Cancer Institute (# HHSN261200800001E) to JDL; and the Bill and Melinda Gates Foundation grant OPP1107409.
- Published
- 2022
- Full Text
- View/download PDF
23. Rectal Acquisition of Simian Immunodeficiency Virus (SIV) SIVmac239 Infection despite Vaccine-Induced Immune Responses against the Entire SIV Proteome
- Author
-
Jeffrey D. Lifson, Patricia L. Earl, Bernard Moss, Dennis R. Burton, Eva G. Rakasz, Lucas Gonzalez-Nieto, Georg F. Bischof, Mauricio A. Martins, Matthias Pauthner, Nuria Pedreño-Lopez, Michael J. Ricciardi, Christine M. Dang, David I. Watkins, Christopher L. Parks, Ronald C. Desrosiers, and Varian K. Bailey
- Subjects
Modified vaccinia Ankara ,Proteome ,animal diseases ,viruses ,Immunology ,Immunization, Secondary ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Antibodies, Viral ,Microbiology ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,HIV vaccine ,Antigens, Viral ,030304 developmental biology ,0303 health sciences ,biology ,Vaccination ,Immunity ,SAIDS Vaccines ,virus diseases ,Simian immunodeficiency virus ,Viral Load ,Vaccine efficacy ,biology.organism_classification ,medicine.disease ,Antibodies, Neutralizing ,Macaca mulatta ,Vesicular stomatitis virus ,Insect Science ,Simian Immunodeficiency Virus ,030215 immunology - Abstract
Given the complex biology of human immunodeficiency virus (HIV) and its remarkable capacity to evade host immune responses, HIV vaccine efficacy may benefit from the induction of both humoral and cellular immune responses of maximal breadth, potency, and longevity. Guided by this rationale, we set out to develop an immunization protocol aimed at maximizing the induction of anti-Envelope (anti-Env) antibodies and CD8(+) T cells targeting non-Env epitopes in rhesus macaques (RMs). Our approach was to deliver the entire simian immunodeficiency virus (SIV) proteome by serial vaccinations. To that end, 12 RMs were vaccinated over 81 weeks with DNA, modified vaccinia Ankara (MVA), vesicular stomatitis virus (VSV), adenovirus type 5 (Ad5), rhesus monkey rhadinovirus (RRV), and DNA again. Both the RRV and the final DNA boosters delivered a near-full-length SIVmac239 genome capable of assembling noninfectious SIV particles and inducing T-cell responses against all nine SIV proteins. Compared to previous SIV vaccine trials, the present DNA-MVA-VSV-Ad5-RRV-DNA regimen resulted in comparable levels of Env-binding antibodies and SIV-specific CD8(+) T-cells. Interestingly, one vaccinee developed low titers of neutralizing antibodies (NAbs) against SIVmac239, a tier 3 virus. Following repeated intrarectal marginal-dose challenges with SIVmac239, vaccinees were not protected from SIV acquisition but manifested partial control of viremia. Strikingly, the animal with the low-titer vaccine-induced anti-SIVmac239 NAb response acquired infection after the first SIVmac239 exposure. Collectively, these results highlight the difficulties in eliciting protective immunity against immunodeficiency virus infection. IMPORTANCE Our results are relevant to HIV vaccine development efforts because they suggest that increasing the number of booster immunizations or delivering additional viral antigens may not necessarily improve vaccine efficacy against immunodeficiency virus infection.
- Published
- 2020
24. Cytomegaloviral determinants of CD8+ T cell programming and RhCMV/SIV vaccine efficacy
- Author
-
Justin M. Greene, Renee G. Espinosa Trethewy, Abigail B. Ventura, Husam Taher, Brian Berkemeier, Luke S. Uebelhoer, Jay A. Nelson, Finn Grey, Colette M. Hughes, Rebecca Shoemaker, Daniel Malouli, Courtney R. Papen, William J. Bosche, Louis J. Picker, David Morrow, Meaghan H. Hancock, Roxanne M. Gilbride, Michael Hull, Michael K. Axthelm, Julia C. Ford, Paul T. Edlefsen, Jonah B. Sacha, Daniel N. Streblow, Kurt T. Randall, Jason Shao, Scott G. Hansen, Jeffrey D. Lifson, Kelli Oswald, Matthew R. McArdle, and Klaus Früh
- Subjects
viruses ,T cell ,Immunology ,virus diseases ,General Medicine ,Simian immunodeficiency virus ,Biology ,Vaccine efficacy ,medicine.disease_cause ,Major histocompatibility complex ,Virology ,medicine.anatomical_structure ,medicine ,biology.protein ,Cytotoxic T cell ,Vector (molecular biology) ,Gene ,CD8 - Abstract
Simian immunodeficiency virus (SIV) insert-expressing, 68-1 Rhesus Cytomegalovirus (RhCMV/SIV) vectors elicit major histocompatibility complex (MHC)-E- and -II-restricted, SIV-specific CD8+ T cell responses, but the basis of these unconventional responses and their contribution to demonstrated vaccine efficacy against SIV challenge in the rhesus monkeys (RMs) has not been characterized. We demonstrate that these unconventional responses resulted from a chance genetic rearrangement in 68-1 RhCMV that abrogated the function of eight distinct immunomodulatory gene products encoded in two RhCMV genomic regions (Rh157.5/.4 and Rh158-161). Differential repair of these genes with either RhCMV-derived or orthologous human CMV (HCMV)-derived sequences (UL128/130; UL146/147) leads to either of two distinct CD8+ T cell response types – MHC-Ia-restricted-only, or a mix of MHC-II- and MHC-Ia-restricted CD8+ T cells. Despite response magnitude and functional differentiation being similar to RhCMV 68-1, neither alternative response type mediated protection against SIV challenge. These findings implicate MHC-E-restricted CD8+ T cell responses as mediators of anti-SIV efficacy and indicate that translation of RhCMV/SIV vector efficacy to humans will likely require deletion of all the genes that inhibit these responses from the HCMV/HIV vector.One-sentence summaryEight genes in two spatially distinct RhCMV gene regions control induction of unconventionally restricted CD8+ T cell responses and the efficacy of RhCMV/SIV vaccine vectors against SIV challenge.
- Published
- 2020
- Full Text
- View/download PDF
25. IL-21 and IFNα therapy rescues terminally differentiated NK cells and limits SIV reservoir in ART-treated macaques
- Author
-
Steven E. Bosinger, Jeffrey D. Lifson, Kirk Easley, Francois Villinger, Hong Wang, Philippe Rascle, Luca Micci, Guido Silvestri, Neeta Shenvi, Gregory K. Tharp, Nicolas Huot, Cristin M. Galardi, Colin T. King, Michaela Müller-Trutwin, Justin L. Harper, Amit A. Upadhyay, Beatrice Jacquelin, Mirko Paiardini, Emory University [Atlanta, GA], HIV, Inflammation et persistance, Institut Pasteur [Paris], Université Paris Diderot, Sorbonne Paris Cité, Paris, France, Université Paris Diderot - Paris 7 (UPD7), University of North Carolina [Chapel Hill] (UNC), University of North Carolina System (UNC), ViiV Healthcare US, ViiV Healthcare [Brentford, UK], University of Louisiana, Frederick National Laboratory for Cancer Research (FNLCR), Emory University School of Medicine, This work was supported by the NIAID, NIH under award number R01AI116379 to M. P. and R01AI143457 to M. M.-T. Support for this work was also provided by ANRS and the Fondation J. Beytout to M. M.-T., NCRR, NIH award 5R24RR016988 to F. V. and the Resource for Nonhuman Primate Immune Reagents, ORIP/OD award P51OD011132 to YNPRC, and NCI, NIH award HHSN261200800001E and 75N91019D00024 to Leidos Biomedical Research. P.R. was recipient of a PhD fellowship from the University Paris Diderot, Sorbonne Paris Cité., HIV, Inflammation et persistance - HIV, Inflammation and Persistence, and Institut Pasteur [Paris] (IP)
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,animal diseases ,Science ,[SDV]Life Sciences [q-bio] ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,General Physics and Astronomy ,Alpha interferon ,Inflammation ,Lymphocyte Activation ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology ,Article ,Natural killer cell ,Natural killer cell differentiation ,03 medical and health sciences ,Interferon-gamma ,0302 clinical medicine ,Interferon ,medicine ,Animals ,Viremia ,Lymph node ,Multidisciplinary ,biology ,Interleukins ,virus diseases ,General Chemistry ,Viral host response ,Viral Load ,biology.organism_classification ,Virology ,Macaca mulatta ,3. Good health ,Killer Cells, Natural ,Rhesus macaque ,030104 developmental biology ,medicine.anatomical_structure ,Anti-Retroviral Agents ,030220 oncology & carcinogenesis ,Female ,Simian Immunodeficiency Virus ,African Green Monkey ,Immunotherapy ,medicine.symptom ,medicine.drug ,HIV infections - Abstract
Unlike HIV infection, which progresses to AIDS absent suppressive anti-retroviral therapy, nonpathogenic infections in natural hosts, such African green monkeys, are characterized by a lack of gut microbial translocation and robust secondary lymphoid natural killer cell responses resulting in an absence of chronic inflammation and limited SIV dissemination in lymph node B-cell follicles. Here we report, using the pathogenic model of antiretroviral therapy-treated, SIV-infected rhesus macaques that sequential interleukin-21 and interferon alpha therapy generate terminally differentiated blood natural killer cells (NKG2a/clowCD16+) with potent human leukocyte antigen-E-restricted activity in response to SIV envelope peptides. This is in contrast to control macaques, where less differentiated, interferon gamma-producing natural killer cells predominate. The frequency and activity of terminally differentiated NKG2a/clowCD16+ natural killer cells correlates with a reduction of replication-competent SIV in lymph node during antiretroviral therapy and time to viral rebound following analytical treatment interruption. These data demonstrate that African green monkey-like natural killer cell differentiation profiles can be rescued in rhesus macaques to promote viral clearance in tissues., Infection of African green monkeys with SIV is associated with reduced pathogenicity. Here the authors explore the requirement of differentiated NK cell populations in a pathogenic Rhesus macaque model of SIV infection and show administration of IL-21 and IFNα rescues terminally differentiated NK cells, similarly to what found in African green monkeys, and limits the SIV reservoir in antiretroviral therapy treated macaques.
- Published
- 2020
- Full Text
- View/download PDF
26. SMAC Mimetic Plus Triple-Combination Bispecific HIVxCD3 Retargeting Molecules in SHIV.C.CH505-Infected, Antiretroviral Therapy-Suppressed Rhesus Macaques
- Author
-
Jeffrey D. Lifson, Guido Ferrari, Guido Silvestri, Maud Mavigner, Chevaughn Waller, Ann Chahroudi, Chia-Ying K Lam, Shan Liang, Richard M. Dunham, Katharine J. Bar, Marina Tuyishime, Amir Dashti, George M. Shaw, Thomas H. Vanderford, Jeffrey L. Nordstrom, Nils Schoof, and David M. Margolis
- Subjects
CD4-Positive T-Lymphocytes ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,HIV Infections ,Inhibitor of apoptosis ,medicine.disease_cause ,Antibodies, Viral ,Virus Replication ,Microbiology ,Inhibitor of Apoptosis Proteins ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,Latency (engineering) ,030304 developmental biology ,0303 health sciences ,biology ,Immunogenicity ,NF-kappa B ,Simian immunodeficiency virus ,Viral Load ,medicine.disease ,Antibodies, Neutralizing ,Macaca mulatta ,Virus Latency ,Anti-Retroviral Agents ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Insect Science ,Alkynes ,biology.protein ,HIV-1 ,Female ,Simian Immunodeficiency Virus ,Antibody ,Viral load ,human activities ,Oligopeptides ,Reassortant Viruses - Abstract
The “shock-and-kill” human immunodeficiency virus type 1 (HIV-1) cure strategy involves latency reversal followed by immune-mediated clearance of infected cells. We have previously shown that activation of the noncanonical NF-κB pathway using an inhibitor of apoptosis (IAP), AZD5582, reverses HIV/simian immunodeficiency virus (SIV) latency. Here, we combined AZD5582 with bispecific HIVxCD3 DART molecules to determine the impact of this approach on persistence. Rhesus macaques (RMs) (n = 13) were infected with simian/human immunodeficiency virus SHIV.C.CH505.375H.dCT, and triple antiretroviral therapy (ART) was initiated after 16 weeks. After 42 weeks of ART, 8 RMs received a cocktail of 3 HIVxCD3 DART molecules having human A32, 7B2, or PGT145 anti-HIV-1 envelope (Env) specificities paired with a human anti-CD3 specificity that is rhesus cross-reactive. The remaining 5 ART-suppressed RMs served as controls. For 10 weeks, a DART molecule cocktail was administered weekly (each molecule at 1 mg/kg of body weight), followed 2 days later by AZD5582 (0.1 mg/kg). DART molecule serum concentrations were well above those considered adequate for redirected killing activity against Env-expressing target cells but began to decline after 3 to 6 weekly doses, coincident with the development of antidrug antibodies (ADAs) against each of the DART molecules. The combination of AZD5582 and the DART molecule cocktail did not increase on-ART viremia or cell-associated SHIV RNA in CD4(+) T cells and did not reduce the viral reservoir size in animals on ART. The lack of latency reversal in the model used in this study may be related to low pre-ART viral loads (median
- Published
- 2020
27. Mucosal immune stimulation with HSV-2 and polyICLC boosts control of viremia in SIVΔNef vaccinated rhesus macaques with breakthrough SIV infection
- Author
-
Thilo Brill, James Blanchard, Olga Mizenina, Brooke Grasperge, Melissa Robbiani, Meropi Aravantinou, Christine Timmons, Jeffrey D. Lifson, Nina Derby, Andres M. Salazar, Ines Frank, Agegnehu Gettie, and Jessica Kenney
- Subjects
Attenuated vaccine ,business.industry ,T cell ,Breakthrough infection ,Viremia ,medicine.disease ,Vaccine efficacy ,Vaccination ,medicine.anatomical_structure ,Immunology ,medicine ,Cytotoxic T cell ,HIV vaccine ,business - Abstract
Development of an effective human immunodeficiency virus (HIV) vaccine is among the highest priorities in the biomedical research agenda. Adjuvants enhance vaccine efficacy, but in the case of HIV, strong or inappropriate immune activation may undermine protection by increasing HIV susceptibility. Co-infection with immunomodulatory pathogens may also impact vaccine efficacy. In the rhesus macaque rectal SIVΔNef live attenuated vaccine model, we utilized a low virulence HSV-2 infection and the double-stranded RNA viral mimic polyICLC as tools to probe the effects of distinct types of immune activation on HIV vaccine efficacy and explore novel correlates of protection from wild type SIV. Rectally administered HSV-2 and polyICLC impacted the protection conferred by mucosal SIVΔNef vaccination by favoring partial protection in animals with breakthrough infection following virulent SIV challenge (“Controllers”). However, SIVΔNef persistence in blood and tissues did not predict protection in this rectal immunization and challenge model. Non-controllers had similar SIVΔNef viremia as completely protected macaques, and while they tended to have less replication competent SIVΔNef in lymph nodes, controllers had no recoverable virus in the lymph nodes. Non-controllers differed from protected macaques immunologically by having a greater frequency of pro-inflammatory CXCR3+CCR6+ CD4 T cells in blood and a monofunctional IFNγ-dominant CD8 T cell response in lymph nodes. Controller phenotype was associated with heightened IFNα production during acute SIV infection and a greater frequency of CXCR5+ CD4 T cells in blood pre-challenge despite a lower frequency of cells with the T follicular helper (Tfh) cell phenotype in blood and lymph nodes. Our results establish novel correlates of immunological control of SIV infection while reinforcing the potential importance of T cell functionality and location in SIVΔNef efficacy. Moreover, this work highlights that triggering of mucosal immunity can aid mucosal vaccine strategies rather than undermine protection.AUTHOR SUMMARYAn efficacious HIV vaccine is essential to contain the HIV pandemic. Vaccine-mediated protection from HIV may be either enhanced or obstructed by mucosal immune activation; thus, the impact of adjuvants and underlying co-infections that lead to immune activation needs to be evaluated. Using the SIV macaque model, we set out to study the impact of underlying infection with HSV-2 or treatment with the adjuvant polyICLC on rectal immunization with the live attenuated vaccine SIVΔNef. We found that neither stimulus impacted complete protection from SIV; however, the combination of HSV-2 and polyICLC improved control of infection in animals that were not completely protected. Compared with non-controller macaques, controllers had less inflammatory T cells before SIV challenge as well as greater gene expression of IFNα and more functional SIV-specific T cells after infection. The results add to our understanding of the mechanisms of SIVΔNef protection and demonstrate that mucosal immune activation does not necessarily undermine protection in mucosal vaccination against HIV.
- Published
- 2020
- Full Text
- View/download PDF
28. Commentary: Derivation of Simian Tropic HIV-1 Infectious Clone Reveals Virus Adaptation to a New Host
- Author
-
Melissa Kane, Jeffrey D. Lifson, Brandon F. Keele, Robert J. Gifford, Alice Raymond, Theodora Hatziioannou, Paul D. Bieniasz, Steven J. Soll, Gregory Q. Del Prete, Dennis Voronin, Fabian Schmidt, Vineet N. KewalRamani, and Christine M. Fennessey
- Subjects
Microbiology (medical) ,viruses ,Immunology ,Human immunodeficiency virus (HIV) ,Clone (cell biology) ,Adaptation, Biological ,lcsh:QR1-502 ,HIV Infections ,Simian ,medicine.disease_cause ,Virus Replication ,Microbiology ,Virus ,Host Specificity ,lcsh:Microbiology ,Evolution, Molecular ,rhesus macaques ,Cellular and Infection Microbiology ,medicine ,Animals ,Cells, Cultured ,pig-tailed macaques ,vif gene ,Multidisciplinary ,biology ,Host (biology) ,General Commentary ,adaptive mutations ,Pigtail macaque ,Biological Sciences ,biology.organism_classification ,Virology ,Phenotype ,Disease Models, Animal ,Infectious Diseases ,Capsid ,Lentivirus ,HIV-1 ,Simian Immunodeficiency Virus ,Capsid Proteins ,Adaptation ,Macaca nemestrina ,macaque-tropic HIV-1 ,SIVmac - Abstract
To replicate in a new host, lentiviruses must adapt to exploit required host factors and evade species-specific antiviral proteins. Understanding how host protein variation drives lentivirus adaptation allowed us to expand the host range of HIV-1 to pigtail macaques. We have previously derived a viral swarm (in the blood of infected animals) that can cause AIDS in this new host. To further exploit this reagent, we generated infectious molecular clones (IMCs) from the viral swarm. We identified clones with high replicative capacity in pigtail peripheral blood mononuclear cells (PBMC) in vitro and used in vivo replication to select an individual IMC, named stHIV-A19 (for simian tropic HIV-1 clone A19), which recapitulated the phenotype obtained with the viral swarm. Adaptation of HIV-1 in macaques led to the acquisition of amino acid changes in viral proteins, such as capsid (CA), that are rarely seen in HIV-1–infected humans. Using stHIV-A19, we show that these CA changes confer a partial resistance to the host cell inhibitor Mx2 from pigtail macaques, but that complete resistance is associated with a fitness defect. Adaptation of HIV-1 to a new host will lead to a more accurate animal model and a better understanding of virus–host interactions.
- Published
- 2020
- Full Text
- View/download PDF
29. Long-Term Delivery of an Anti-SIV Monoclonal Antibody With AAV
- Author
-
Ronald C. Desrosiers, Sebastian P. Fuchs, Jeffrey D. Lifson, José M. Martinez-Navio, Guangping Gao, Eva G. Rakasz, and Desiree E. Mendes
- Subjects
0301 basic medicine ,Time Factors ,Genetic enhancement ,viruses ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,Antibodies, Viral ,law.invention ,0302 clinical medicine ,law ,Immunology and Allergy ,Original Research ,Disease Resistance ,AIDS Vaccines ,Antibodies, Monoclonal ,Dependovirus ,Viral Load ,gene therapy ,long-term expression ,3. Good health ,AAV vector ,Recombinant DNA ,Female ,Simian Immunodeficiency Virus ,immunotherapy ,prophylaxis ,Antibody ,Viral load ,lcsh:Immunologic diseases. Allergy ,medicine.drug_class ,Genetic Vectors ,Immunology ,Enzyme-Linked Immunosorbent Assay ,Biology ,Gp41 ,Monoclonal antibody ,Virus ,03 medical and health sciences ,medicine ,Animals ,Humans ,rhesus monkeys ,broadly neutralizing antibodies ,Virology ,Macaca mulatta ,030104 developmental biology ,Pepscan ,biology.protein ,HIV-1 ,lcsh:RC581-607 ,030215 immunology ,HIV/SIV cure - Abstract
Long-term delivery of anti-HIV monoclonal antibodies using adeno-associated virus (AAV) holds promise for the prevention and treatment of HIV infection. We previously reported that after receiving a single administration of AAV vector coding for anti-SIV antibody 5L7, monkey 84-05 achieved high levels of AAV-delivered 5L7 IgG1 in vivo which conferred sterile protection against six successive, escalating dose, intravenous challenges with highly infectious, highly pathogenic SIVmac239, including a final challenge with 10 animal infectious doses (1). Here we report that monkey 84-05 has successfully maintained 240-350 μg/ml of anti-SIV antibody 5L7 for over 6 years. Approximately 2% of the circulating IgG in this monkey is this one monoclonal antibody. This monkey generated little or no anti-drug antibodies (ADA) to the AAV-delivered antibody for the duration of the study. Due to the nature of the high-dose challenge used and in order to rule out a potential low-level infection not detected by regular viral loads, we have used ultrasensitive techniques to detect cell-associated viral DNA and RNA in PBMCs from this animal. In addition, we have tested serum from 84-05 by ELISA against overlapping peptides spanning the whole envelope sequence for SIVmac239 (PepScan) and against recombinant p27 and gp41 proteins. No reactivity has been detected in the ELISAs indicating the absence of naturally arising anti-SIV antibodies; moreover, the ultrasensitive cell-associated viral tests yielded no positive reaction. We conclude that macaque 84-05 was effectively protected and remained uninfected. Our data show that durable, continuous antibody expression can be achieved after one single administration of AAV and support the potential for lifelong protection against HIV from a single vector administration.
- Published
- 2020
- Full Text
- View/download PDF
30. Immunotherapy with DNA vaccine and live attenuated rubella/SIV gag vectors plus early ART can prevent SIVmac251 viral rebound in acutely infected rhesus macaques
- Author
-
Konstantin Virnik, Aaron Scanlan, Mark A. Lewis, Margherita Rosati, Gabrielle Walsh, Frances Dayton, Ruth M. Ruprecht, Yvonne J. Bryson, Barbara K. Felber, Jeffrey D. Lifson, Kate E. Broderick, Ira Berkower, and Alexei Medvedev
- Subjects
0301 basic medicine ,RNA viruses ,Viral Diseases ,Time Factors ,Physiology ,medicine.medical_treatment ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Monkeys ,Virus Replication ,Pathology and Laboratory Medicine ,Biochemistry ,Rubella vaccine ,White Blood Cells ,0302 clinical medicine ,Immunodeficiency Viruses ,Animal Cells ,Immune Physiology ,Vaccines, DNA ,Medicine and Health Sciences ,030212 general & internal medicine ,Mammals ,Vaccines ,Multidisciplinary ,Immune System Proteins ,T Cells ,Viral Vaccine ,SAIDS Vaccines ,Eukaryota ,Combined Modality Therapy ,3. Good health ,Virus Latency ,Treatment Outcome ,Infectious Diseases ,SIV ,Medical Microbiology ,Viral Pathogens ,Vertebrates ,Viruses ,Medicine ,Drug Therapy, Combination ,Simian Immunodeficiency Virus ,Immunotherapy ,Cellular Types ,Pathogens ,Viral load ,Macaque ,medicine.drug ,Plasmids ,Research Article ,Primates ,Infectious Disease Control ,Anti-HIV Agents ,Immune Cells ,Science ,Genetic Vectors ,Immunology ,Gene Products, gag ,Cytotoxic T cells ,Vaccines, Attenuated ,Rubella ,Microbiology ,Virus ,Drug Administration Schedule ,Antibodies ,DNA vaccination ,03 medical and health sciences ,Old World monkeys ,Retroviruses ,medicine ,Animals ,Humans ,Microbial Pathogens ,Acquired Immunodeficiency Syndrome ,Blood Cells ,Biology and life sciences ,business.industry ,Lentivirus ,Organisms ,Proteins ,Cell Biology ,medicine.disease ,Virology ,Macaca mulatta ,Disease Models, Animal ,030104 developmental biology ,Immunization ,Amniotes ,Clinical Immunology ,Clinical Medicine ,business ,Rubella virus - Abstract
Anti-retroviral therapy (ART) has been highly successful in controlling HIV replication, reducing viral burden, and preventing both progression to AIDS and viral transmission. Yet, ART alone cannot cure the infection. Even after years of successful therapy, ART withdrawal leads inevitably to viral rebound within a few weeks or months. Our hypothesis: effective therapy must control both the replicating virus pool and the reactivatable latent viral reservoir. To do this, we have combined ART and immunotherapy to attack both viral pools simultaneously. The vaccine regimen consisted of DNA vaccine expressing SIV Gag, followed by a boost with live attenuated rubella/gag vectors. The vectors grow well in rhesus macaques, and they are potent immunogens when used in a prime and boost strategy. We infected rhesus macaques by high dose mucosal challenge with virulent SIVmac251 and waited three days to allow viral dissemination and establishment of a reactivatable viral reservoir before starting ART. While on ART, the control group received control DNA and empty rubella vaccine, while the immunotherapy group received DNA/gag prime, followed by boosts with rubella vectors expressing SIV gag over 27 weeks. Both groups had a vaccine "take" to rubella, and the vaccine group developed antibodies and T cells specific for Gag. Five weeks after the last immunization, we stopped ART and monitored virus rebound. All four control animals eventually had a viral rebound, and two were euthanized for AIDS. One control macaque did not rebound until 2 years after ART release. In contrast, there was only one viral rebound in the vaccine group. Three out of four vaccinees had no viral rebound, even after CD8 depletion, and they remain in drug-free viral remission more than 2.5 years later. The strategy of early ART combined with immunotherapy can produce a sustained SIV remission in macaques and may be relevant for immunotherapy of HIV in humans.
- Published
- 2020
31. Ultrasensitive Immunoassay for Simian Immunodeficiency Virus p27CA
- Author
-
Anitha Vijayagopalan, Siddhartha A. K. Datta, Adrienne E. Swanstrom, Gregory Q. Del Prete, Kelli Oswald, Bonnie J. Howell, Jeffrey D. Lifson, Rebecca Shoemaker, Julian W. Bess, Elena Chertova, Guoxin Wu, Robert J. Gorelick, and Brandon F. Keele
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Cart ,viruses ,Definitive Therapy ,030106 microbiology ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Gene Products, gag ,medicine.disease_cause ,Sensitivity and Specificity ,Persistence (computer science) ,03 medical and health sciences ,Virology ,Replication (statistics) ,medicine ,Antiretroviral treatment ,Animals ,Immunoassay ,medicine.diagnostic_test ,business.industry ,virus diseases ,Simian immunodeficiency virus ,Macaca mulatta ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Anti-Retroviral Agents ,Viral replication ,RNA, Viral ,Simian Immunodeficiency Virus ,Virus Activation ,business - Abstract
Although effective for suppressing viral replication, combination antiretroviral treatment (cART) does not represent definitive therapy for HIV infection due to persistence of replication-competent viral reservoirs. The advent of effective cART regimens for simian immunodeficiency virus (SIV)-infected nonhuman primates (NHP) has enabled the development of relevant models for studying viral reservoirs and intervention strategies targeting them. Viral reservoir measurements are crucial for such studies but are problematic. Quantitative polymerase chain reaction (PCR) assays overestimate the size of the replication competent viral reservoir, as not all detected viral genomes are intact. Quantitative viral outgrowth assays measure replication competence, but they suffer from limited precision and dynamic range, and require large numbers of cells. Ex vivo virus induction assays to detect cells harboring inducible virus represent an experimental middle ground, but detection of inducible viral RNA in such assays does not necessarily indicate production of virions, while detection of more immunologically relevant viral proteins, including p27(CA), by conventional enzyme-linked immunosorbent assays (ELISA) lacks sensitivity. An ultrasensitive digital SIV Gag p27 assay was developed, which is 100-fold more sensitive than a conventional ELISA. In ex vivo virus induction assays, the quantification of SIV Gag p27 produced by stimulated CD4+ T cells from rhesus macaques receiving cART enabled earlier and more sensitive detection than conventional ELISA-based approaches and was highly correlated with SIV RNA, as measured by quantitative reverse transcription PCR. This ultrasensitive p27 assay provides a new tool to assess ongoing replication and reactivation of infectious virus from reservoirs in SIV-infected NHP.
- Published
- 2018
- Full Text
- View/download PDF
32. Defining total-body AIDS-virus burden with implications for curative strategies
- Author
-
Gregory J. Beilman, Jeffrey D. Lifson, Francis Ssali, Jordan Schoephoerster, Steven G. Deeks, Samuel P. Callisto, Claire Deleage, Courtney V. Fletcher, Jacob D. Estes, Jacob Jasurda, Torfi Hoskuldsson, Thomas E. Schmidt, Joseph M. McCune, Jared Schuster, Michael Hafertepe, Alexander Khoruts, Cissy Kityo, Gregory Q. Del Prete, Thomas Reimann, Peter J. Southern, Timothy W. Schacker, Paul A. Luciw, Ashley T. Haase, Katherine Perkey, Krystelle Nganou Makamdop, Jodi Anderson, Hope Pearson, Jeffrey G. Chipman, Liang Shang, Louise A. Swainson, Daniel C. Douek, and Stephen W. Wietgrefe
- Subjects
0301 basic medicine ,Drug ,Anti-HIV Agents ,Lymphoid Tissue ,media_common.quotation_subject ,030106 microbiology ,HIV Infections ,Article ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Persistence (computer science) ,03 medical and health sciences ,Acquired immunodeficiency syndrome (AIDS) ,medicine ,Humans ,media_common ,business.industry ,HIV ,RNA ,General Medicine ,Viral Load ,medicine.disease ,Virology ,030104 developmental biology ,Lymphatic system ,Viral replication ,DNA, Viral ,Immunology ,RNA, Viral ,business ,Viral load - Abstract
In the quest for a functional cure or eradication of HIV infection, we need to know how large the reservoirs are from which infection rebounds when treatment is interrupted. To that end, we quantified SIV and HIV tissue burdens in tissues of infected non-human primates and lymphoid tissue (LT) biopsies from infected humans. Before antiretroviral therapy (ART), LTs harbor more than 98 percent of the SIV RNA+ and DNA+ cells. While ART substantially reduced their numbers, vRNA+ cells were still detectable and their persistence was associated with relatively low drug concentrations in LT compared to peripheral blood. Prolonged ART also reduced the level of SIV and HIV-DNA+ cells, but the estimated size of the residual tissue burden of 108 vDNA+ cells that potentially harbor replication competent proviruses, along with the evidence for continuing virus production in LT despite ART, identify two important sources for rebound following treatment interruption. The large sizes of these tissue reservoirs underscore the challenges in developing “HIV cure” strategies that target multiple sources of virus production.
- Published
- 2017
- Full Text
- View/download PDF
33. Frequent Use of Khat, an Amphetamine-Like Substance, as a Risk Factor for Poor Adherence and Lost to Follow-Up Among Patients New to HIV Care in Ethiopia
- Author
-
Behailu Dagne, Sale Workneh, Rose Hilk, Desalegn Admassu Ayana, Zenebe Melaku, Hiwot Tekle Waktola, Tibebe Shenie, Lucy Slater, Ken C. Winters, Alan R. Lifson, and Lemlem Bezabih
- Subjects
Adult ,Male ,medicine.medical_specialty ,Substance-Related Disorders ,Epidemiology ,Immunology ,Human immunodeficiency virus (HIV) ,HIV Infections ,Catha ,medicine.disease_cause ,Poor adherence ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Khat ,Surveys and Questionnaires ,Virology ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Risk factor ,Lost to follow-up ,Amphetamine ,030505 public health ,biology ,business.industry ,biology.organism_classification ,Frequent use ,Infectious Diseases ,Quartile ,Patient Compliance ,Female ,Lost to Follow-Up ,Ethiopia ,0305 other medical science ,business ,medicine.drug - Abstract
Khat, a plant native to East Africa, has amphetamine-like psychoactive constituents, and is a potential risk factor for HIV infection. Chronic use can cause cognitive impairment and other mental disorders, raising concerns about effects on retention and adherence with HIV care. During 2013–2014, 322 Ethiopian patients newly enrolled at HIV clinics in Dire Dawa and Harar were surveyed about khat use and prospectively followed for 1 year; 9% died, 18% transferred care to other clinics, and 22% were lost to follow-up (LTFU) (no clinic visit for >3 months). Of 248 patients who received a 12-month follow-up survey, 37% used khat in the year after enrollment, with a median use of 60 h in a typical month. Those using khat ≥60 h/month (median among users) were more likely than others to be LTFU (31% vs. 16%, p = .014); those using khat ≥150 h/month (upper quartile) had 44% LTFU rates versus 16% for others (p = .002). Complete 3-day adherence (taking all doses) of antiretroviral therapy was reported by 77% of those using khat ≥60 h/month versus 95% of all others (p
- Published
- 2017
- Full Text
- View/download PDF
34. An intravaginal ring that releases three antiviral agents and a contraceptive blocks SHIV-RT infection, reduces HSV-2 shedding, and suppresses hormonal cycling in rhesus macaques
- Author
-
Meropi Aravantinou, Michael L. Cooney, Keith Levendosky, James Blanchard, Brooke Grasperge, Jessica Kenney, José A. Fernández-Romero, Jolanta Wilk, Samantha Seidor, Shweta R. Ugaonkar, Shimin Zhang, Michael Piatak, Olga Mizenina, Melissa Robbiani, Larisa Kizima, Thomas M. Zydowsky, Jeffrey D. Lifson, Aixa Rodriguez, Nina Derby, Agegnehu Gettie, and Asa Wesenberg
- Subjects
0301 basic medicine ,Pyridines ,Herpesvirus 2, Human ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Zinc Acetate ,Pharmaceutical Science ,Alphapapillomavirus ,Carrageenan ,medicine.disease_cause ,Contraceptive Agents, Female ,Urea ,Medicine ,Levonorgestrel ,Immunodeficiency ,Transmission (medicine) ,virus diseases ,HSV-2 ,Virus Shedding ,3. Good health ,Contraception ,Vaginal Creams, Foams, and Jellies ,Original Article ,Drug Therapy, Combination ,Female ,medicine.drug ,HPV ,030106 microbiology ,Antiviral Agents ,03 medical and health sciences ,In vivo ,Animals ,Humans ,Viral shedding ,Menstrual Cycle ,business.industry ,HIV ,Contraceptive Devices, Female ,Herpes Simplex ,Simian immunodeficiency virus ,medicine.disease ,Multipurpose prevention technology ,Macaca mulatta ,Virology ,Reverse transcriptase ,Disease Models, Animal ,Intravaginal ring ,030104 developmental biology ,Immunology ,business ,Hormone - Abstract
Women globally need access to multipurpose prevention technologies (MPTs) that prevent human immunodeficiency virus (HIV), sexually transmitted infections that increase HIV acquisition/transmission risk, and unintended pregnancy. Seeking an MPT with activity against HIV, herpes simplex virus-2 (HSV-2), and human papillomavirus (HPV), we developed a prototype intravaginal ring (IVR), the MZCL IVR, which released the antiviral agents MIV-150, zinc acetate, and carrageenan (MZC for short) and the contraceptive levonorgestrel (LNG). Previously, we showed that an MZC gel has potent activity against immunodeficiency viruses, HSV-2, and HPV and that the MZCL (MZC with LNG) IVR releases all four components in macaques in vivo at levels associated with efficacy. Vaginal fluid from treated macaques has in vitro activity against HIV, HSV-2, and HPV. Herein, we assessed the ability of the MZCL IVR to protect macaques against repeated co-challenge with HSV-2 and SHIV-RT (simian immunodeficiency virus [SIV] containing the reverse transcriptase gene from HIV) and prevent hormonal cycling. We evaluated in vivo drug release in co-challenged macaques by measuring drug levels in blood and vaginal fluid and residual drug levels in used IVRs. The MZCL IVR significantly prevented SHIV-RT infection, reduced HSV-2 vaginal shedding, and prevented cycling. No non-nucleoside HIV reverse transcriptase inhibitor (NNRTI)-resistant SHIV was detected in macaques that became infected after continuous exposure to MZC from the IVR. Macaques wearing the MZCL IVR also had carrageenan levels in vaginal fluid expected to protect from HPV (extrapolated from mice) and LNG levels in blood associated with contraceptive efficacy. The MZCL IVR is a promising MPT candidate that warrants further development.
- Published
- 2017
- Full Text
- View/download PDF
35. Improved quality of life with immediate versus deferred initiation of antiretroviral therapy in early asymptomatic HIV infection
- Author
-
Eric Florence, Eileen Denning, Fabian Chen, Sounkalo Dao, Jesús Sanz, Edward M. Gardner, Sean Emery, Richard Kaplan, Nicole Engen, Birgit Grund, Alan R. Lifson, and Catherine L. Carey
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Visual analogue scale ,Immunology ,MEDLINE ,Human immunodeficiency virus (HIV) ,HIV Infections ,medicine.disease_cause ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Randomized controlled trial ,law ,Antiretroviral Therapy, Highly Active ,Surveys and Questionnaires ,Secondary Prevention ,Humans ,Immunology and Allergy ,Medicine ,030212 general & internal medicine ,business.industry ,030112 virology ,Antiretroviral therapy ,Mental health ,humanities ,CD4 Lymphocyte Count ,Clinical trial ,Treatment Outcome ,Infectious Diseases ,Anti-Retroviral Agents ,Quality of Life ,Physical therapy ,Female ,business - Abstract
Objective: To determine if immediate compared to deferred initiation of antiretroviral therapy (ART) in healthy persons living with HIV had a more favorable impact on health-related quality of life (QOL), or self-assessed physical, mental, and overall health status. Design: QOL was measured in the Strategic Timing of Antiretroviral Therapy study, which randomized healthy ART-naive persons living with HIV with CD4 cell counts above 500 cells/μl from 35 countries to immediate versus deferred ART. Methods: At baseline, months 4 and 12, then annually, participants completed a visual analog scale (VAS) for perceived current health and the Short-Form 12-Item Health Survey version 2 from which the following were computed: general health perception; physical component summary (PCS); and mental component summary (MCS); the VAS and general health were rated from 0 (lowest) to 100 (highest). Results: QOL at study entry was high (mean scores: VAS=80.9, general health=72.5, PCS53.7, MCS=48.2). Over a mean follow-up of 3 years, changes in all QOL measures favored the immediate group (P < 0.001); estimated differences were as follows: VAS=1.9, general health=3.6, PCS=0.8, MCS=0.9. When QOL changes were assessed across various demographic and clinical subgroups, treatment differences continued to favor the immediate group. QOL was poorer in those experiencing primary outcomes; however, when excluding those with primary events, results remained favorable for immediate ART recipients. Conclusion: In an international randomized trial in ART-naive participants with above 500 CD4 cells/μl, there were modest but significant improvements in self-assessed QOL among those initiating ART immediately compared to deferring treatment, supporting patient-perceived health benefits of initiating ART as soon as possible after an HIV diagnosis.
- Published
- 2017
- Full Text
- View/download PDF
36. Shocking HIV out of hiding
- Author
-
Steven E. Bosinger, Gavin C. Sampey, Zhang Wang, Jean-Pierre Routy, Robert G. Ferris, Rae Ann Spagnuolo, Edward P. Browne, Brandon F. Keele, Greg K. Tharp, Christopher C. Nixon, Nils Schoof, Christine M. Fennessey, Chandrav De, Corinne G. Cammon, Rachel A. Cleary, David Favre, Nancie M. Archin, J. Victor Garcia, Jennifer Deutsch, Angela Wahl, Sean Maguire, Hasse Walum, Thomas H. Vanderford, Cameron Mattingly, David M. Margolis, Baolin Liao, Nathaniel J. Schramm, Richard M. Dunham, Amit A. Upadhyay, Jessica H. Brehm, Cristin M. Galardi, Saintedym Wills, Corbin D. Jones, Maud Mavigner, Sherrie Jean, Jeffrey D. Lifson, Matt Kanke, Guido Silvestri, Phong T. Ho, Alyssa D. Brooks, Ann Chahroudi, David M. Irlbeck, and Shane D. Falcinelli
- Subjects
0301 basic medicine ,History ,Computer science ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Human immunodeficiency virus (HIV) ,HIV Infections ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Mice ,0302 clinical medicine ,Virus latency ,Drug Discovery ,Multidisciplinary ,NF-kappa B ,virus diseases ,General Medicine ,Hedgehog signaling pathway ,Virus Latency ,Computer Science Applications ,medicine.anatomical_structure ,Anti-Retroviral Agents ,Alkynes ,030220 oncology & carcinogenesis ,Simian Immunodeficiency Virus ,Lymph ,Oligopeptides ,MEDLINE ,Article ,Virus ,Education ,03 medical and health sciences ,In vivo ,medicine ,Animals ,Humans ,Latency (engineering) ,Pharmacology ,business.industry ,Simian immunodeficiency virus ,NFKB1 ,medicine.disease ,Macaca mulatta ,Virology ,030104 developmental biology ,Immunology ,HIV-1 ,Bone marrow ,business ,030215 immunology - Abstract
Long-lasting, latently infected resting CD4+ T cells are the greatest obstacle to obtaining a cure for HIV infection, as these cells can persist despite decades of treatment with antiretroviral therapy (ART). Estimates indicate that more than 70 years of continuous, fully suppressive ART are needed to eliminate the HIV reservoir1. Alternatively, induction of HIV from its latent state could accelerate the decrease in the reservoir, thus reducing the time to eradication. Previous attempts to reactivate latent HIV in preclinical animal models and in clinical trials have measured HIV induction in the peripheral blood with minimal focus on tissue reservoirs and have had limited effect2–9. Here we show that activation of the non-canonical NF-κB signalling pathway by AZD5582 results in the induction of HIV and SIV RNA expression in the blood and tissues of ART-suppressed bone-marrow–liver–thymus (BLT) humanized mice and rhesus macaques infected with HIV and SIV, respectively. Analysis of resting CD4+ T cells from tissues after AZD5582 treatment revealed increased SIV RNA expression in the lymph nodes of macaques and robust induction of HIV in almost all tissues analysed in humanized mice, including the lymph nodes, thymus, bone marrow, liver and lung. This promising approach to latency reversal—in combination with appropriate tools for systemic clearance of persistent HIV infection—greatly increases opportunities for HIV eradication. Activation of the non-canonical NF-κB signalling pathway by AZD5582 results in the induction of HIV and SIV RNA expression in the blood and tissues of antiretroviral-therapy-treated humanized mice and rhesus macaques.
- Published
- 2020
- Full Text
- View/download PDF
37. In Vivo Validation of the Viral Barcoding of Simian Immunodeficiency Virus SIVmac239 and the Development of New Barcoded SIV and Subtype B and C Simian-Human Immunodeficiency Viruses
- Author
-
Sirish Khanal, Gregory Q. Del Prete, Brandon F. Keele, Abigail Thorpe, Louis J. Picker, Julian W. Bess, Taina T. Immonen, Sean P. O’Brien, Miles P. Davenport, Adrienne E. Swanstrom, Rodman Smith, Carolyn Reid, Christine M. Fennessey, Afam A. Okoye, and Jeffrey D. Lifson
- Subjects
0303 health sciences ,viruses ,Immunology ,Population structure ,Human immunodeficiency virus (HIV) ,Simian immunodeficiency virus ,Biology ,Simian ,medicine.disease_cause ,biology.organism_classification ,Microbiology ,Genome ,Virology ,Virus ,Deep sequencing ,03 medical and health sciences ,0302 clinical medicine ,Genetic Diversity and Evolution ,In vivo ,Insect Science ,medicine ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Genetically barcoded viral populations are powerful tools for evaluating the overall viral population structure as well as assessing the dynamics and evolution of individual lineages in vivo over time. Barcoded viruses are generated by inserting a small, genetically unique tag into the viral genome, which is retained in progeny virus. We recently reported barcoding the well-characterized molecular clone simian immunodeficiency virus (SIV) SIVmac239, resulting in a synthetic swarm (SIVmac239M) containing approximately 10,000 distinct viral clonotypes for which all genetic differences were within a 34-base barcode that could be tracked using next-generation deep sequencing. Here, we assessed the population size, distribution, and authenticity of individual viral clonotypes within this synthetic swarm using samples from 120 rhesus macaques infected intravenously. The number of replicating barcodes in plasma correlated with the infectious inoculum dose, and the primary viral growth rate was similar in all infected animals regardless of the inoculum size. Overall, 97% of detectable clonotypes in the viral stock were identified in the plasma of at least one infected animal. Additionally, we prepared a second-generation barcoded SIVmac239 stock (SIVmac239M2) with over 16 times the number of barcoded variants of the original stock and an additional barcoded stock with suboptimal nucleotides corrected (SIVmac239Opt5M). We also generated four barcoded stocks from subtype B and C simian-human immunodeficiency virus (SHIV) clones. These new SHIV clones may be particularly valuable models to evaluate Env-targeting approaches to study viral transmission or viral reservoir clearance. Overall, this work further establishes the reliability of the barcoded virus approach and highlights the feasibility of adapting this technique to other viral clones. IMPORTANCE We recently developed and published a description of a barcoded simian immunodeficiency virus that has a short random sequence inserted directly into the viral genome. This allows for the tracking of individual viral lineages with high fidelity and ultradeep sensitivity. This virus was used to infect 120 rhesus macaques, and we report here the analysis of the barcodes of these animals during primary infection. We found that the vast majority of barcodes were functional in vivo. We then expanded the barcoding approach in a second-generation SIVmac239 stock (SIVmac239M2) with over 16 times the number of barcoded variants of the original stock and a barcoded stock of SIVmac239Opt5M whose sequence had 5 changes from the wild-type SIVmac239 sequence. We also generated 4 barcoded stocks from subtype B and C SHIV clones each containing a human immunodeficiency virus (HIV) type 1 envelope. These virus models are functional and can be useful for studying viral transmission and HIV cure/reservoir research.
- Published
- 2019
38. Evaluating the Intactness of Persistent Viral Genomes in Simian Immunodeficiency Virus-Infected Rhesus Macaques after Initiating Antiretroviral Therapy within One Year of Infection
- Author
-
Brandon F. Keele, Gregory Q. Del Prete, Samuel Long, Robert J. Gorelick, Christine M. Fennessey, Yuan Li, Sean P. O’Brien, Jeffrey D. Lifson, Carolyn Reid, and Laura Newman
- Subjects
CD4-Positive T-Lymphocytes ,Cart ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,Genome, Viral ,Biology ,Virus Replication ,medicine.disease_cause ,Microbiology ,Genome ,Virus ,Frameshift mutation ,03 medical and health sciences ,0302 clinical medicine ,Antiretroviral Therapy, Highly Active ,Raltegravir Potassium ,Virology ,medicine ,Animals ,Emtricitabine ,Tenofovir ,030304 developmental biology ,0303 health sciences ,Whole Genome Sequencing ,Point mutation ,Genomics ,Viral Load ,Simian immunodeficiency virus ,medicine.disease ,Macaca mulatta ,Chronic infection ,Anti-Retroviral Agents ,Genetic Diversity and Evolution ,Insect Science ,DNA, Viral ,Mutation ,RNA, Viral ,Simian Immunodeficiency Virus ,030217 neurology & neurosurgery - Abstract
The major obstacle to more-definitive treatment for HIV infection is the early establishment of virus that persists despite long-term combination antiretroviral therapy (cART) and can cause recrudescent viremia if cART is interrupted. Previous studies of HIV DNA that persists despite cART indicated that only a small fraction of persistent viral sequences was intact. Experimental simian immunodeficiency virus (SIV) infections of nonhuman primates (NHPs) are essential models for testing interventions designed to reduce the viral reservoir. We studied the viral genomic integrity of virus that persists during cART under conditions typical of many NHP reservoir studies, specifically with cART started within 1 year postinfection and continued for at least 9 months. The fraction of persistent DNA in SIV-infected NHPs starting cART during acute or chronic infection was assessed with a multiamplicon, real-time PCR assay designed to analyze locations that are regularly spaced across the viral genome to maximize coverage (collectively referred to as “tile assay”) combined with near-full-length (nFL) single-genome sequencing. The tile assay is used to rapidly screen for major deletions, with nFL sequence analysis used to identify additional potentially inactivating mutations. Peripheral blood mononuclear cells (PBMC) from animals started on cART within 1 month of infection, sampled at least 9 months after cART initiation, contained at least 80% intact genomes, whereas those from animals started on cART 1 year postinfection and treated for 1 year contained intact genomes only 47% of the time. The most common defect identified was large deletions, with the remaining defects caused by APOBEC-mediated mutations, frameshift mutations, and inactivating point mutations. Overall, this approach can be used to assess the intactness of persistent viral DNA in NHPs. IMPORTANCE Molecularly defining the viral reservoir that persists despite antiretroviral therapy and that can lead to rebound viremia if antiviral therapy is removed is critical for testing interventions aimed at reducing this reservoir. In HIV infection in humans with delayed treatment initiation and extended treatment duration, persistent viral DNA has been shown to be dominated by nonfunctional genomes. Using multiple real-time PCR assays across the genome combined with near-full-genome sequencing, we defined SIV genetic integrity after 9 to 18 months of combination antiretroviral therapy in rhesus macaques starting therapy within 1 year of infection. In the animals starting therapy within a month of infection, the vast majority of persistent DNA was intact and presumptively functional. Starting therapy within 1 year increased the nonintact fraction of persistent viral DNA. The approach described here allows rapid screening of viral intactness and is a valuable tool for assessing the efficacy of novel reservoir-reducing interventions.
- Published
- 2019
- Full Text
- View/download PDF
39. How long is long-term? Delivery of anti-HIV antibodies using AAV vector
- Author
-
G. Gao, Sebastian P. Fuchs, D. Mendes, Eva G. Rakasz, Jeffrey D. Lifson, José M. Martinez-Navio, and Ronald C. Desrosiers
- Subjects
biology ,Epidemiology ,Anti hiv ,business.industry ,Immunology ,Public Health, Environmental and Occupational Health ,Virology ,Microbiology ,QR1-502 ,Infectious Diseases ,biology.protein ,Medicine ,Term delivery ,Vector (molecular biology) ,Antibody ,Public aspects of medicine ,RA1-1270 ,business - Published
- 2019
40. The latency reversal activity of the SMAC mimetic AZD5582 in ART-suppressed SIV-infected rhesus macaques is potentiated by CD8a cell depletion
- Author
-
Jeffrey D. Lifson, Brandon F. Keele, Alyssa D. Brooks, Thomas H. Vanderford, Cameron Mattingly, Guido Silvestri, Ann Chahroudi, Maud Mavigner, Richard M. Dunham, and David M. Margolis
- Subjects
Epidemiology ,Immunology ,Cell ,Public Health, Environmental and Occupational Health ,Biology ,Smac mimetics ,Microbiology ,QR1-502 ,CD8A ,Cell biology ,Infectious Diseases ,medicine.anatomical_structure ,Virology ,medicine ,Latency (engineering) ,Public aspects of medicine ,RA1-1270 - Published
- 2019
41. Evaluating latency reactivation synergies between the bromodomain inhibitor iBET-151 and the SMAC mimetic AZD5582 in SIV-infected macaques on ART
- Author
-
Afam A. Okoye, B.E. Randall, Jeremy Smedley, Richard M. Dunham, R. Lum, Shane D. Falcinelli, Louis J. Picker, Jeffrey D. Lifson, B. Varco-Merth, and Yoshinori Fukazawa
- Subjects
Epidemiology ,Immunology ,Public Health, Environmental and Occupational Health ,Biology ,Smac mimetics ,Microbiology ,QR1-502 ,Bromodomain ,Infectious Diseases ,Virology ,Cancer research ,Latency (engineering) ,Public aspects of medicine ,RA1-1270 - Published
- 2019
42. Barcoded viruses facilitate tracking changes to the composition of the rebound-competent reservoir
- Author
-
Jeffrey D. Lifson, Christine M. Fennessey, Brandon F. Keele, and T. Immonen
- Subjects
Infectious Diseases ,Epidemiology ,Virology ,Immunology ,Public Health, Environmental and Occupational Health ,Biology ,Public aspects of medicine ,RA1-1270 ,Tracking (particle physics) ,Biological system ,Composition (language) ,Microbiology ,QR1-502 - Published
- 2019
43. Preferential Small Intestine Homing and Persistence of CD8 T Cells in Rhesus Macaques Achieved by Molecularly Engineered Expression of CCR9 and Reduced Ex Vivo Manipulation
- Author
-
Lori V. Coren, Sumiti Jain, David E. Ott, Matthew W. Breed, Gregory Q. Del Prete, Matthew T. Trivett, Adrienne E. Swanstrom, Eugene V. Barsov, Jeffrey D. Lifson, Claire Deleage, Brenna J. Hill, Joshua A. Kramer, and James D. Burke
- Subjects
0303 health sciences ,Adoptive cell transfer ,Immunology ,CD28 ,Simian immunodeficiency virus ,Biology ,medicine.disease_cause ,Microbiology ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,030220 oncology & carcinogenesis ,Virology ,Insect Science ,medicine ,Cytotoxic T cell ,CD8 ,Ex vivo ,030304 developmental biology ,Homing (hematopoietic) - Abstract
Adoptive cell transfer (ACT) is a powerful experimental approach to directly study T-cell-mediated immunity in vivo. In the rhesus macaque AIDS virus model, infusing simian immunodeficiency virus (SIV)-infected animals with CD8 T cells engineered to express anti-SIV T-cell receptor specificities enables direct experimentation to better understand antiviral T-cell immunity in vivo. Limiting factors in ACT experiments include suboptimal trafficking to, and poor persistence in, the secondary lymphoid tissues targeted by AIDS viruses. Previously, we redirected CD8 T cells to B-cell follicles by ectopic expression of the CXCR5 homing protein. Here, we modify peripheral blood mononuclear cell (PBMC)-derived CD8 T cells to express the CCR9 chemokine receptor, which induces preferential homing of the engineered cells to the small intestine, a site of intense early AIDS virus replication and pathology in rhesus macaques. Additionally, we increase in vivo persistence and overall systemic distribution of infused CD8 T cells, especially in secondary lymphoid tissues, by minimizing ex vivo culture/manipulation, thereby avoiding the loss of CD28+/CD95+ central memory T cells by differentiation in culture. These proof-of-principle results establish the feasibility of preferentially localizing PBMC-derived CD8 T cells to the small intestine and enables the direct experimental ACT-based assessment of the potential role of the quality and timing of effective antiviral CD8 T-cell responses to inhibit viral infection and subsequent replication in small intestine CD4 T cells. More broadly, these results support the engineered expression of homing proteins to direct CD8 T cells to target tissues as a means for both experimental and potential therapeutic advances in T-cell immunotherapies, including cancer. IMPORTANCEAdoptive cell transfer (ACT) of T cells engineered with antigen-specific effector properties can deliver targeted immune responses against malignancies and infectious diseases. Current T-cell-based therapeutic ACT relies on circulatory distribution to deliver engineered T cells to their targets, an approach which has proven effective for some leukemias but provided only limited efficacy against solid tumors. Here, engineered expression of the CCR9 homing receptor redirected CD8 T cells to the small intestine in rhesus macaque ACT experiments. Targeted homing of engineered T-cell immunotherapies holds promise to increase the effectiveness of adoptively transferred cells in both experimental and clinical settings.
- Published
- 2019
- Full Text
- View/download PDF
44. Erratum for the Report: 'Elevated HLA - A expression impairs HIV control through inhibition of NKG2A-expressing cells' by V. Ramsuran, V. Naranbhai, A. Horowitz, Y. Qi, M. P. Martin, Y. Yuki, X. Gao, V. Walker-Sperling, G. Q. Del Prete, D. K. Schneider, J. D. Lifson, J. Fellay, S. G. Deeks, J. N. Martin, J. J. Goedert, S. M. Wolinsky, N. L. Michael, G. D. Kirk, S. Buchbinder, D. Haas, T. Ndung’u, P. Goulder, P. Parham, B. D. Walker, J. M. Carlson, M. Carrington
- Author
-
Susan Buchbinder, Peter Parham, Gregory D. Kirk, Steven M. Wolinsky, Philip J. R. Goulder, Mary Carrington, Ying Qi, Amir Horowitz, Yuko Yuki, Douglas K. Schneider, Nelson L. Michael, Vivek Naranbhai, David W. Haas, Thumbi Ndung'u, James J. Goedert, Jonathan M. Carlson, Bruce D. Walker, Jeffrey D. Lifson, Jeffrey N. Martin, Steven G. Deeks, Veron Ramsuran, Maureen P. Martin, Xiaojiang Gao, Jacques Fellay, G.Q. Del Prete, and Victoria Walker-Sperling
- Subjects
Multidisciplinary ,Expression (architecture) ,Immunology ,Human immunodeficiency virus (HIV) ,medicine ,Biology ,medicine.disease_cause ,HLA-A - Published
- 2019
- Full Text
- View/download PDF
45. Fingolimod treatment at ART initiation delays SIV rebound after ART interruption
- Author
-
Constantinos Petrovas, Justin L. Harper, I. Shim, Barbara Cervasi, H. Wang, Sadia Samer, Claire Deleage, Jeffrey D. Lifson, C. King, Arnold Reynaldi, M. Paiardini, Maria Pino, K. Nguyen, Kartika Padhan, Miles P. Davenport, and Michael M. Lederman
- Subjects
Oncology ,medicine.medical_specialty ,Epidemiology ,business.industry ,Art initiation ,Immunology ,Public Health, Environmental and Occupational Health ,Fingolimod ,Microbiology ,QR1-502 ,Infectious Diseases ,Virology ,Internal medicine ,medicine ,Public aspects of medicine ,RA1-1270 ,business ,medicine.drug - Published
- 2019
46. Clonal expansion of SIV-infected cells in macaques on antiretroviral therapy is similar to that of HIV-infected cells in humans
- Author
-
Shuang Guo, Andrea L. Ferris, Adrienne E. Swanstrom, Gregory Q. Del Prete, David Wells, Stephen H. Hughes, Jeffrey D. Lifson, John M. Coffin, and Xiaolin Wu
- Subjects
CD4-Positive T-Lymphocytes ,RNA viruses ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,Monkeys ,Virus Replication ,Pathology and Laboratory Medicine ,Macaque ,White Blood Cells ,Immunodeficiency Viruses ,Animal Cells ,Medicine and Health Sciences ,Genomic library ,Biology (General) ,Lymph node ,Mammals ,0303 health sciences ,biology ,T Cells ,030302 biochemistry & molecular biology ,Eukaryota ,virus diseases ,Genomics ,Viral Load ,medicine.anatomical_structure ,Anti-Retroviral Agents ,SIV ,Medical Microbiology ,Viral Pathogens ,Vertebrates ,Viruses ,Infectious diseases ,Simian Immunodeficiency Virus ,Pathogens ,Cellular Types ,Research Article ,Primates ,QH301-705.5 ,Virus Integration ,Immune Cells ,Immunology ,Spleen ,Viral diseases ,In Vitro Techniques ,Research and Analysis Methods ,Microbiology ,Virus ,Human Genomics ,03 medical and health sciences ,Virology ,biology.animal ,Old World monkeys ,Retroviruses ,medicine ,Genetics ,Animals ,Humans ,Molecular Biology Techniques ,Gene ,Microbial Pathogens ,Molecular Biology ,030304 developmental biology ,Disease Reservoirs ,Cloning ,Blood Cells ,Host Microbial Interactions ,Biology and life sciences ,Lentivirus ,Organisms ,HIV ,Computational Biology ,Cell Biology ,RC581-607 ,Genome Analysis ,Genomic Libraries ,Macaca mulatta ,In vitro ,Amniotes ,Parasitology ,Immunologic diseases. Allergy - Abstract
Clonal expansion of HIV infected cells plays an important role in the formation and persistence of the reservoir that allows the virus to persist, in DNA form, despite effective antiretroviral therapy. We used integration site analysis to ask if there is a similar clonal expansion of SIV infected cells in macaques. We show that the distribution of HIV and SIV integration sites in vitro is similar and that both viruses preferentially integrate in many of the same genes. We obtained approximately 8000 integration sites from blood samples taken from SIV-infected macaques prior to the initiation of ART, and from blood, spleen, and lymph node samples taken at necropsy. Seven clones were identified in the pre-ART samples; one persisted for a year on ART. An additional 100 clones were found only in on-ART samples; a number of these clones were found in more than one tissue. The timing and extent of clonal expansion of SIV-infected cells in macaques and HIV-infected cells in humans is quite similar. This suggests that SIV-infected macaques represent a useful model of the clonal expansion of HIV infected cells in humans that can be used to evaluate strategies intended to control or eradicate the viral reservoir., Author summary Although antiretroviral therapy (ART) effectively blocks HIV replication, infected people are not cured. As a part of its normal replication cycle, HIV inserts (integrates) a DNA copy of its genome into the genome of infected host cells, which allows the virus to persist as long as the infected cells survive. Not only can these infected cells survive, they can grow and divide, increasing the numbers of infected cells without viral replication. The ability of the infected cells to proliferate plays an important role in maintaining the numbers of infected cells (and the infection) in people on successful therapy. However, there are some important experiments that cannot easily be done with samples that can be obtained from HIV infected people. SIV infected macaques are often used as a model to do experiments that cannot be done in HIV infected people. We show here that the distribution of HIV and SIV integration sites is similar, and that, in infected macaques, the timing and extent of the proliferation of SIV infected cells is also quite similar to HIV infected cells in humans. This shows that the SIV/macaque system can be used to model the clonal expansion of HIV infected cells.
- Published
- 2019
47. TLR7 agonist administration to SIV-infected macaques receiving early initiated cART does not induce plasma viremia
- Author
-
Tiffany Barnes, Jeffrey D. Lifson, Matthew W. Breed, Charles M. Trubey, Rodney Wiles, Jacob Kiser, Randy Fast, Cathi Pyle, Joshua A. Kramer, Vicky Coalter, Mukta Nag, Kelli Oswald, Rebecca Kiser, W. Gregory Alvord, William J. Bosche, Gregory Q. Del Prete, Joseph Hesselgesser, Tyler Malys, Claire Deleage, Yuan Li, Lorna Silipino, Brian Berkemeier, Michael Hull, Elizabeth Chipriano, Jacob D. Estes, Romas Geleziunas, James A. Thomas, and Adam Wiles
- Subjects
Male ,0301 basic medicine ,Agonist ,Cart ,medicine.drug_class ,T cell ,Population ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,CD8-Positive T-Lymphocytes ,Peripheral blood mononuclear cell ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,education ,education.field_of_study ,business.industry ,Pteridines ,virus diseases ,General Medicine ,Viral Load ,medicine.disease ,Macaca mulatta ,Up-Regulation ,030104 developmental biology ,medicine.anatomical_structure ,Anti-Retroviral Agents ,Toll-Like Receptor 7 ,030220 oncology & carcinogenesis ,Immunology ,Cytokines ,RNA, Viral ,Drug Therapy, Combination ,business ,CD8 ,Research Article - Abstract
Reduction/elimination of HIV-1 reservoirs that persist despite combination antiretroviral therapy (cART) will likely require induction of viral expression by residual infected cells and enhanced clearance of these cells. TLR7 agonists have potential to mediate these activities. We evaluated immunologic and virologic effects of repeated doses of the TLR7 agonist GS-9620 in SIV-infected rhesus macaques receiving cART, which was initiated at 13 days after infection and was continued for 75 weeks prior to GS-9620 administration. During cART, GS-9620 induced transient upregulation of IFN-stimulated genes in blood and tissues, increases in plasma cytokines, and changes in immune cell population activation and phenotypes but did not result in measurable increases in plasma viremia or viral RNA–to–viral DNA ratio in PBMCs or tissues nor decreases in viral DNA in PBMC or tissues. SIV-specific CD8(+) T cell responses, negligible prior to GS-9620 treatment, were not measurably boosted by treatment; a second course of GS-9620 administration overlapping with later cART discontinuation was associated with increased CD8(+) T cell responses during viral recrudescence. These results confirm and extend evidence for GS-9620–mediated enhancement of antiviral immune responses in SIV-infected macaques but suggest that GS-9620–mediated viral induction may depend critically on the timing of initiation and duration of cART and resulting characteristics of viral reservoirs.
- Published
- 2019
- Full Text
- View/download PDF
48. PD-1 Blockade and TLR7 Activation Lack Therapeutic Benefit in Chronic Simian Immunodeficiency Virus-Infected Macaques on Antiretroviral Therapy
- Author
-
Joseph Hesselgesser, Lance Stapleton, Agneta von Gegerfelt, Mark Nagel, Adele Wang, Magdeleine Hung, Jeffrey D. Lifson, Elena Bekerman, Romas Geleziunas, Brian A. Carr, and Hanne Andersen Elyard
- Subjects
Male ,simian immunodeficiency virus ,Viral pathogenesis ,T cell ,medicine.medical_treatment ,viruses ,Programmed Cell Death 1 Receptor ,Simian Acquired Immunodeficiency Syndrome ,Context (language use) ,medicine.disease_cause ,Antiviral Agents ,Antibodies ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Pharmacology (medical) ,030304 developmental biology ,Pharmacology ,0303 health sciences ,biology ,human immunodeficiency virus ,business.industry ,Pteridines ,Immunotherapy ,Simian immunodeficiency virus ,Viral Load ,Flow Cytometry ,Macaca mulatta ,Blockade ,Toll-like receptors ,Infectious Diseases ,medicine.anatomical_structure ,Anti-Retroviral Agents ,Toll-Like Receptor 7 ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,immunotherapy ,Antibody ,business - Abstract
Antiretroviral therapy (ART) limits human immunodeficiency virus 1 (HIV-1) replication but does not eliminate the long-lived reservoir established shortly after viral acquisition. A successful HIV cure intervention necessitates either elimination or generation of long-term immune control of the persistent viral reservoir. Immune modulating strategies in conjunction with ART hold promise for achieving cure by inducing viral antigen expression and augmenting infected cell killing., Antiretroviral therapy (ART) limits human immunodeficiency virus 1 (HIV-1) replication but does not eliminate the long-lived reservoir established shortly after viral acquisition. A successful HIV cure intervention necessitates either elimination or generation of long-term immune control of the persistent viral reservoir. Immune modulating strategies in conjunction with ART hold promise for achieving cure by inducing viral antigen expression and augmenting infected cell killing. Programmed death-1 (PD-1) blockade is a potential means to both activate and eliminate the latent reservoir by restoring exhausted T cell function. We assessed the therapeutic efficacy of PD-1 blockade, Toll-like receptor 7 (TLR7) activation with the agonist vesatolimod, or a combination of the two agents in chronically simian immunodeficiency virus (SIV)-infected macaques suppressed with ART for more than 2 years. Despite achieving extended anti-PD-1 antibody plasma exposure and TLR7-dependent immune activation after multiple administrations, neither individual treatment nor the combination resulted in changes to viral rebound kinetics following ART interruption or reduction in the SIV reservoir size. Our data in the context of other reports demonstrating improved viral control upon PD-1 blockade suggest that its therapeutic utility may be restricted to specific experimental conditions or treatment times during viral pathogenesis.
- Published
- 2019
49. Short Communication: Ultrasensitive Immunoassay for Assessing Residual Simian-Tropic HIV in Nonhuman Primate Models of AIDS
- Author
-
Robert J. Gorelick, Gregory Q. Del Prete, Jeffrey D. Lifson, Theodora Hatziioannou, Adrienne E. Swanstrom, Paul D. Bieniasz, and Alison Jacques
- Subjects
0301 basic medicine ,Cart ,Viral protein ,viruses ,Immunology ,HIV Core Protein p24 ,Simian Acquired Immunodeficiency Syndrome ,medicine.disease_cause ,Virus Replication ,Sensitivity and Specificity ,03 medical and health sciences ,0302 clinical medicine ,Virology ,medicine ,Animals ,030212 general & internal medicine ,Disease Reservoirs ,Immunoassay ,medicine.diagnostic_test ,biology ,virus diseases ,Pigtail macaque ,Viral Load ,biology.organism_classification ,Macaca mulatta ,Reverse transcription polymerase chain reaction ,030104 developmental biology ,Infectious Diseases ,Viral replication ,Models, Animal ,HIV-1 ,Simian Immunodeficiency Virus ,Virus Activation ,Cell culture assays ,Ex vivo - Abstract
Persistence of replication-competent viral reservoirs during infection remains a barrier to HIV cure, despite the ability of combination antiretroviral therapy (cART) to effectively suppress viral replication. Simian-tropic HIV (stHIV) is a minimally chimeric HIV-1 that is comprised of 94% HIV-1 sequence, contains HIV-1 drug and immunologic targets, and is capable of replicating to high levels and causing authentic HIV-like pathogenesis leading to clinical AIDS in pigtail macaques. Suppression of stHIV replication by cART provides a model for study of viral reservoirs and HIV-specific intervention strategies targeting them. Accurate measurement of reservoir size is crucial for evaluating the effect of any such intervention strategies. Although there are a variety of assays that allow for indirect monitoring of viral reservoir size ex vivo, they each quantify a different aspect of viral reservoirs, and are characterized by conceptual and/or technical limitations. Measurement of viral protein in ex vivo cell culture assays captures the immunologically relevant viral-antigen producing component of the reservoir. This study demonstrates the utility of an ultrasensitive digital HIV Gag p24 immunoassay, which enabled earlier, and more sensitive detection of viral protein in culture supernatants from stimulated CD4+ T cells from stHIV-infected pigtail macaques receiving cART compared with conventional enzyme-linked immunosorbent assay. Protein measurements were highly correlated with cell-free stHIV RNA, as measured by quantitative reverse transcription polymerase chain reaction. This ultrasensitive p24 assay can be used to complement other reservoir measurement tools to assess ongoing replication and reactivation of infectious virus from reservoirs in stHIV-infected pigtail macaques.
- Published
- 2019
50. Advanced HIV Disease among Males and Females Initiating HIV Care in Rural Ethiopia
- Author
-
Alan R. Lifson, Keith J. Horvath, Richard F. MacLehose, Sale Workneh, Anne Sites, Rose Hilk, Abera Hailemichael, and Tibebe Shenie
- Subjects
sub-Saharan Africa ,Adult ,Male ,Rural Population ,0301 basic medicine ,medicine.medical_specialty ,Delayed Diagnosis ,Immunology ,Human immunodeficiency virus (HIV) ,HIV Infections ,Dermatology ,Disease ,CD4 count ,World Health Organization ,medicine.disease_cause ,lcsh:RC870-923 ,World health ,Young Adult ,03 medical and health sciences ,Sex Factors ,0302 clinical medicine ,Weight loss ,advanced HIV disease ,Surveys and Questionnaires ,Internal medicine ,gender ,medicine ,Humans ,Mass index ,030212 general & internal medicine ,Hiv treatment ,2. Zero hunger ,business.industry ,virus diseases ,HIV ,Middle Aged ,lcsh:Diseases of the genitourinary system. Urology ,030112 virology ,CD4 Lymphocyte Count ,3. Good health ,Rural ethiopia ,Infectious Diseases ,Anti-Retroviral Agents ,Original Article ,Female ,Ethiopia ,medicine.symptom ,business ,Hiv disease - Abstract
Despite recommendations for rapidly initiating HIV treatment, many persons in sub-Saharan Africa present to care with advanced HIV disease. Baseline survey and clinical data were collected on 1799 adults newly enrolling at 32 district hospitals and local health HIV clinics in rural Ethiopia. Among those with complete HIV disease information, advanced HIV disease (defined as CD4 count 3 or World Health Organization [WHO] HIV clinical stage III or IV disease) was present in 66% of males and 56% of females ( P < .001). Males (compared to females) had lower CD4 counts (287 cells/mm3 versus 345 cells/mm3), lower body mass index (19.3 kg/m2 versus 20.2 kg/m2), and more WHO stage III or IV disease (46% versus 37%), ( P < .001). Men reported more chronic diarrhea, fevers, cough, pain, fatigue, and weight loss ( P < .05). Most initiating care in this resource-limited setting had advanced HIV disease. Men had poorer health status, supporting the importance of earlier diagnosis, linkage to care, and initiation of antiretroviral therapy.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.